1
|
Ma Y, Fung V, VanKeulen-Miller R, Tiwade PB, Narasipura EA, Gill NA, Fenton OS. A Metabolite Co-Delivery Strategy to Improve mRNA Lipid Nanoparticle Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26202-26215. [PMID: 40274610 DOI: 10.1021/acsami.4c22969] [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: 04/26/2025]
Abstract
Lipid nanoparticles (LNPs) effectively protect mRNA and facilitate its entry into target cells for protein synthesis. Despite these successes, cellular entry alone may not be enough for optimal protein expression, as mRNA translation also depends on the availability of essential metabolites, including metabolic energy sources, coenzymes, and amino acids. Without adequate metabolites, mRNA translation may be less efficient, potentially leading to higher dosing requirements or poorer therapeutic outcomes for LNP therapies. To address this, we develop a metabolite co-delivery strategy by encapsulating essential metabolites within mRNA LNPs, hypothesizing that our approach can uniformly improve mRNA delivery. Instead of adding a fifth component to the organic phase, our strategy involves mixing the metabolite with the mRNA payload in the aqueous phase, while maintaining the molar ratio of the components in the organic phase during LNP formulation. We verify our approach in vitro and in vivo, highlighting the broad applicability of our strategy through mechanism and efficacy studies across multiple cell lines, and physiological conditions, such as normoxia (i.e., 21% oxygen), hypoxia (i.e., 1% oxygen), and in mice. Taken collectively, we anticipate that our metabolite co-delivery strategy may serve as a generalizable strategy to enhance in vitro and in vivo protein expression using mRNA LNPs, potentially offering broad applicability for the study and treatment of disease.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Palas B Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan A Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Nicole A Gill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
2
|
Ji Z, He L, Sun M, Lv M, Chen R, Zhao C, Ma L, Cheng J, Qin J, Xu X, Fan Z. Nanoporous Plasmonic Microneedle Arrays Induced High-Efficiency Intracellular Delivery of Metabolism Regulating Protein. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412106. [PMID: 40042405 DOI: 10.1002/smll.202412106] [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: 12/12/2024] [Revised: 02/05/2025] [Indexed: 04/11/2025]
Abstract
Patterned micro/nanostructure arrays have shown the potential to effectively regulate cellular behavior, and their unique microstructure may address the limitations of conventional pore materials, leading to novel phenomena. In this work, a large-area gold micro/nano-array substrate with an average hole of ≈32 nm is designed and extensively screened. Precisely engineered nanopores on the substrate can effectively improve photothermal conversion efficiency, and instant heat dissipation in the absence of laser irradiation. The mesoporous arrays are fabricated by hybrid lithography, offering advantages such as simple processing, high reproducibility, and immense commercial potential. Notably, its heating rate is as rapid as ≈45 K µs-1 at low power levels, with the cooling duration reduced to ≈50 µs after the laser irradiation. Metabolism regulatory proteins such as cytochrome C (CytoC) and β-galactosidase (β-gal) can be efficiently introduced into the U87 cell model without inducing phototoxicity or protein inactivation, maintaining catalytic activity to modulate the cellular metabolic state. This delivery platform based on transient nano-cyclones stimulating cell perturbations can be further expanded through modulated microstructures, such as delivering functional proteins or biomolecules for efficient intracellular regulation, cellular transfection, and in the future application as a potential high-throughput screening tool for clustered regularly interspaced short palindromic repeats (CAR-T) biopharmaceutical and clustered regularly interspaced short palindromic repeats (CRISPR) technologies.
Collapse
Affiliation(s)
- Zhenkai Ji
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| | - Le He
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Min Sun
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| | - Mingchen Lv
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| | - Ran Chen
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| | - Chuanzhen Zhao
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jiajing Cheng
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Jinlong Qin
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| | - Zhen Fan
- Department of Gynecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- School of Materials Science and Engineering, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China
| |
Collapse
|
3
|
Janes ME, Gottlieb AP, Park KS, Acharya S, Bibbey MG, Mitragotri S. Controlling vaccine kinetics using tannic acid for enhanced humoral immunity. J Control Release 2025; 379:135-146. [PMID: 39733913 DOI: 10.1016/j.jconrel.2024.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
Despite the success of global vaccination campaigns, vaccine access in low-resource settings is an ongoing challenge. Subunit vaccines are a well-established and clinically scalable intervention, yet they have achieved limited success for poorly immunogenic antigens such as those associated with SARS-CoV-2. Delivery strategies that promote gradual release of subunit vaccines from the injection site offer the potential to improve humoral immunity by enhancing lymph node exposure, however, clinical implementation of this strategy is challenging due to poor scalability and high costs. Here, we propose an approach that uses the polyphenol tannic acid (TA) as a simple and inexpensive strategy to enhance tissue residence of vaccines and subsequent humoral immunity. We show that TA mediates supramolecular interactions between vaccine components and tissue at the subcutaneous injection site to promote extended retention of protein antigens for over one week. In addition to enhancing the magnitude and duration of vaccine drainage to the lymph nodes, inclusion of TA improved accumulation of activated, antigen-laden monocyte-derived dendritic cells (moDCs), promoting long-lasting humoral immunity against the receptor-binding domain (RBD) of SARS-CoV-2 and variants of concern. This system, termed TAPER (Tannic Acid-Promoted Enhanced Retention) provides various translational advantages including one-pot synthesis, scalability, low cost, and modularity, towards realization of effective and accessible subunit vaccines.
Collapse
Affiliation(s)
- Morgan E Janes
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA; John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Shrinivas Acharya
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Michael Griffith Bibbey
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Allston, MA 02134, USA; Wyss Institute of Biologically Inspired Engineering, Boston, MA 02215, USA.
| |
Collapse
|
4
|
Ming P, Li B, Li Q, Yuan L, Jiang X, Liu Y, Cai R, Zhou P, Lan X, Tao G, Xiao J. Multifunctional sericin-based biomineralized nanoplatforms with immunomodulatory and angio/osteo-genic activity for accelerated bone regeneration in periodontitis. Biomaterials 2025; 314:122885. [PMID: 39423514 DOI: 10.1016/j.biomaterials.2024.122885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Periodontitis is a chronic inflammation caused by dental plaque. It is characterized by the accumulation of excessive reactive oxygen species (ROS) and inflammatory mediators in the periodontal area. This affects the function of host cells, activates osteoclasts, and destroys periodontal tissue. Treatments such as local debridement or antibiotic therapy for ameliorating the overactive inflammatory microenvironment and repairing periodontal tissues are challenging. This paper reports multifunctional nanoplatforms (Se-CuSrHA@EGCG) based on sericin with ROS-scavenging, immunomodulatory, angiogenic, and osteogenic capabilities. The natural protein sericin, derived from silk cocoons, is used in water/oil emulsification and cross-linking processes to create sericin nanoparticles (Se NPs). Numerous binding sites are present on the surface of Se NPs. Ion-doped hydroxyapatite nanoparticles (Se-CuSrHA NPs) can be constructed using the force between positive and negative charges. After mineralization, an antioxidant coating is formed on the surface using polyethyleneimine (PEI)/epigallocatechin gallate (EGCG). Research conducted both in vitro and in vivo demonstrates that Se-CuSrHA@EGCG NPs can efficiently scavenge ROS, regulate macrophage polarization, increase the secretion of anti-inflammatory cytokines, and balance the immune microenvironment. In addition, Se-CuSrHA@EGCG stimulates angiogenesis, inhibits osteoclasts, and accelerates periodontal tissue repair. Therefore, this is a preferable strategy to accelerate bone regeneration in patients with periodontitis.
Collapse
Affiliation(s)
- Piaoye Ming
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China
| | - Bojiang Li
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Qiumei Li
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Lingling Yuan
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xueyu Jiang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yunfei Liu
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Rui Cai
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China; Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Peirong Zhou
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China; Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Gang Tao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China; Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China.
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, 646000, China; Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, 646000, China; Institute of Stomatology, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
5
|
Upadhyaya J, Singh IR, Pun B, Baishya HJ, Kumar S, Joshi SR, Mitra S. Therapeutic Advantages of Nanoparticle-Impregnated Lysozyme Conjugates toward Amyloid-β Fibrillation and Antimicrobial Activity. J Phys Chem B 2025; 129:1274-1288. [PMID: 39812393 DOI: 10.1021/acs.jpcb.4c07261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The interaction of protein with nanoparticles (NPs) of varying shape and/or size boosts our understanding on their bioreactivity and establishes a comprehensive database for use in medicine, diagnosis, and therapeutic applications. The present study explores the interaction between lysozyme (LYZ) and different NPs like graphene oxide (GO) and zinc oxide (ZnO) having various shapes (spherical, 's', and rod-shaped, 'r') and sizes, focusing on their binding dynamics and subsequent effects on both the protein fibrillation and antimicrobial properties. Typically, GO is considered a promising medium due to its apparent inhibition and prolonged lag phase for LYZ fibrillation. However, the present results showed that spherical ZnO NPs (sZnO) offer superior efficacy in modulating fibrillation with an extended lag time of about 158.70 h, further emphasizing the importance of detailed investigation on the nanomaterial characteristics and fibril formation kinetics beyond initial observations. The experimental findings further confirmed a strong correlation between the binding affinity of NPs to the native protein and their effective inhibition of protein denaturation, ultimately preventing fibril formation. Interestingly, the lysozyme nanoconjugates showed intriguing bactericidal effects, as confirmed through the agar plate assay and SEM imaging, over the native protein. Overall, this study shows that appropriate bionanomaterials can exhibit multifunctional properties, which paves the way for a deeper investigation of NP characteristics, ultimately benefiting a wide array of intriguing research.
Collapse
Affiliation(s)
- Jahnabi Upadhyaya
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| | | | - Bishal Pun
- Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong 793022, India
| | - Hirak Jyoti Baishya
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Sugam Kumar
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - S R Joshi
- Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong 793022, India
| | - Sivaprasad Mitra
- Department of Chemistry, North-Eastern Hill University, Shillong 793 022, India
| |
Collapse
|
6
|
Dong P, Qiu H, Wen R, Zou X, Sun X, Yu L, Zhang S, Wu Y, Lan F. Reactive Oxygen and Nitrogen Species - "Nanosweeper" for Rheumatoid Arthritis Theranostics by Macrophage Reprogramming. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70322-70338. [PMID: 39666274 DOI: 10.1021/acsami.4c16772] [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: 12/13/2024]
Abstract
Excessive reactive oxygen and nitrogen species (RONS) accumulation in joints are significant variables that affect the course of rheumatoid arthritis (RA). Scavenging of RONS to remodel macrophage homeostasis is a potentially powerful treatment for RA. Here, a visualized "nanosweeper" by functionalizing ultrasmall Gd/Fe3O4 nanoparticles with thiol-polyethylene glycol-phosphoric acid and 2-(3-(2-aminophenyl)ureido) ethyl methacrylate hydrochloride (APUEMA), namely GIA NPs, can simultaneously scavenge both nitric oxide (NO) and reactive oxygen species (ROS), as well as enhance magnetic resonance imaging (MRI) for the diagnosis and therapy of RA. GIA NPs could not only eliminate NO by reactions between the o-phenylenediamine moieties of APUEMA and NO molecules but also remove ROS by the superoxide peroxidase activities of the Fe3O4 nanoparticles and thiol. In vitro and in vivo experiments revealed that the simultaneous scavenging of NO and ROS strategy inhibited the overactivation of the cyclic guanosine monophosphate/protein kinase G (cGMP/PKG) signaling pathway to reprogram the polarization states of macrophages and interfered with metabolism to alleviate RA. In addition, GIA NPs, as dual-modal nanoprobes for MRI, exhibited the capacity for the early diagnosis of RA lesions and monitoring during the RA treatment process. The visualized "nanosweeper" strategy provides a promising integrated platform for the diagnosis and treatment of RA.
Collapse
Affiliation(s)
- Pingli Dong
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Haoyu Qiu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Rui Wen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiang Zou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiaoqing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Lingzhu Yu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Shiyong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Fang Lan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
7
|
Ma Y, Li S, Lin X, Chen Y. A perspective of lipid nanoparticles for RNA delivery. EXPLORATION (BEIJING, CHINA) 2024; 4:20230147. [PMID: 39713203 PMCID: PMC11655307 DOI: 10.1002/exp.20230147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/07/2024] [Indexed: 12/24/2024]
Abstract
Over the last two decades, lipid nanoparticles (LNPs) have evolved as an effective biocompatible and biodegradable RNA delivery platform in the fields of nanomedicine, biotechnology, and drug delivery. They are novel bionanomaterials that can be used to encapsulate a wide range of biomolecules, such as mRNA, as demonstrated by the current successes of COVID-19 mRNA vaccines. Therefore, it is important to provide a perspective on LNPs for RNA delivery, which further offers useful guidance for researchers who want to work in the RNA-based LNP field. This perspective first summarizes the approaches for the preparation of LNPs, followed by the introduction of the key characterization parameters. Then, the in vitro cell experiments to study LNP performance, including cell selection, cell viability, cellular association/uptake, endosomal escape, and their efficacy, were summarized. Finally, the in vivo animal experiments in the aspects of animal selection, administration, dosing and safety, and their therapeutic efficacy were discussed. The authors hope this perspective can offer valuable guidance to researchers who enter the field of RNA-based LNPs and help them understand the crucial parameters that RNA-based LNPs demand.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Shiyao Li
- School of ScienceRMIT UniversityBundooraVictoriaAustralia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical EngineeringThe University of MelbourneParkvilleVictoriaAustralia
| | - Xin Lin
- Department of Cell BiologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
8
|
Xu L, Fang J, Pan J, Qi H, Yin Y, He Y, Gan X, Li Y, Li Y, Guo J. Zinc finger-inspired peptide-metal-phenolic nanointerface enhances bone-implant integration under bacterial infection microenvironment through immune modulation and osteogenesis promotion. Bioact Mater 2024; 41:564-576. [PMID: 39257672 PMCID: PMC11384338 DOI: 10.1016/j.bioactmat.2024.08.009] [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: 05/10/2024] [Revised: 07/22/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
Orthopedic and dental implantations under bacterial infection microenvironment face significant challenges in achieving high-quality bone-implant integration. Designing implant coatings that incorporate both immune defense and anti-inflammation is difficult in conventional single-functional coatings. We introduce a multifunctional nanointerface using a zinc finger-inspired peptide-metal-phenolic nanocoating, designed to enhance implant osseointegration under such conditions. Abaloparatide (ABL), a second-generation anabolic drug for treating osteoporosis, can be integrated into the design of a zinc-phenolic network constructed on the implant surface (ABL@ZnTA). Importantly, the phenolic-coordinated Zn2+ ions in ABL@ZnTA can act as zinc finger motif to co-stabilize the configuration of ABL through multiple molecular interactions, enabling high bioactivity, high loading capacity (1.36 times), and long-term release (>7 days) of ABL. Our results showed that ABL@ZnTA can modulate macrophage polarization from the pro-inflammatory M1 towards the anti-inflammatory M2 phenotype, promoting immune osteogenesis with increased OCN, ALP, and SOD 1 expression. Furthermore, the ABL@ZnTA significantly reduces inflammatory fibrous tissue encapsulation and enhances the long-term stability of the implants, indicated by enhanced binding strength (6 times) and functional connectivity (1.5-3 times) in the rat bone defect model infected by S. aureus. Overall, our research offers a nano-enabled synergistic strategy that balances infection defense and osteogenesis promotion in orthopedic and dental implantations.
Collapse
Affiliation(s)
- Lin Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiezhou Pan
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Hexu Qi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yun Yin
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Bioproducts Institute, Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
9
|
Dong H, Zhao Y, Li S, Wang Q, Li M, Zhao K, Zhang Z, Shi J, Zhao X, Liu J. An inhalable nanoparticle enabling virulence factor elimination and antibiotics delivery for pneumococcal pneumonia therapy. J Control Release 2024; 375:698-711. [PMID: 39313100 DOI: 10.1016/j.jconrel.2024.09.036] [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: 04/20/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Streptococcus pneumoniae (S. pneumoniae) is a major cause of community-acquired pneumonia. Current standard clinical therapies mainly focus on combating S. pneumoniae through antibiotics. However, the limited delivery of antibiotics and the undetoxified hydrogen peroxide (H2O2) virulence factor secreted by S. pneumoniae impede the therapeutic outcomes. Here we report an inhalable catalase (CAT)-tannic acid (TA) nanoassembly for local antibiotic (levofloxacin) delivery and simultaneously neutralizing the secreted H2O2 virulence factors to treat pneumococcal pneumonia. After aerosol inhalation, the inhalable formulation (denoted as CT@LVX) effectively accumulates in lung tissues through TA-mediated mucoadhesion. CAT can reduce alveolar epithelial cells apoptosis by catalyzing the decomposition of accumulated H2O2 in the infected lung tissues. In synergy with antibiotic LVX-mediated S. pneumoniae elimination, CT@LVX significantly decreases lung injury companied with reduced inflammatory, resulting in 100 % survival of mice with pneumonia. In a clinically isolated S. pneumoniae strain-induced pneumonia mouse model, CT@LVX also shows superior outcomes compared to the traditional antibiotic treatment, highlighting its potential clinical application prospects.
Collapse
Affiliation(s)
- Huiyue Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuxin Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shihong Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Qiongwei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengli Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaikai Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiu Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
10
|
Ma X, Zhang Z, Barba-Bon A, Han D, Qi Z, Ge B, He H, Huang F, Nau WM, Wang X. A small-molecule carrier for the intracellular delivery of a membrane-impermeable protein with retained bioactivity. Proc Natl Acad Sci U S A 2024; 121:e2407515121. [PMID: 39436658 PMCID: PMC11536097 DOI: 10.1073/pnas.2407515121] [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: 04/15/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Intracellular protein delivery has the potential to revolutionize cell-biological research and medicinal therapy, with broad applications in bioimaging, disease treatment, and genome editing. Herein, we demonstrate successful delivery of a functional protein, cytochrome c (CYC), by using a boron cluster anion as molecular carrier of the superchaotropic anion type (B12Br11OPr2-). CYC was delivered into lipid bilayer vesicles as well as living cells, with a cellular uptake ratio approaching 90%. Mechanistic studies showed that CYC was internalized into cells through a permeation pathway directly into the cytoplasm, bypassing endosomal entrapment. Upon carrier-assisted internalization, CYC retained its bioactivity, as reflected by an induced cell apoptosis rate of 25% at low dose (1 µM). This study furbishes a direct protein delivery method by a molecular carrier with high efficiency, confirming the potential of inorganic cluster ions as protein transport vehicles with an extensive range of future cell-biological or biomedical applications.
Collapse
Affiliation(s)
- Xiqi Ma
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Zhixiong Zhang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | | | - Dongxue Han
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Zichun Qi
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Baosheng Ge
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Hua He
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Fang Huang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| | - Werner M. Nau
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
- School of Science, Constructor University, Bremen28759, Germany
| | - Xiaojuan Wang
- College of Chemical Engineering, China University of Petroleum (East China), Qingdao266580, China
| |
Collapse
|
11
|
Liu Y, Wang J, Sun Z. Aromatic Biobased Polymeric Materials Using Plant Polyphenols as Sustainable Alternative Raw Materials: A Review. Polymers (Basel) 2024; 16:2752. [PMID: 39408462 PMCID: PMC11479198 DOI: 10.3390/polym16192752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
In the foreseeable future, the development of petroleum-based polymeric materials may be limited, owing to the gradual consumption of disposable resources and the increasing emphasis on environmental protection policies. Therefore, it is necessary to focus on introducing environmentally friendly renewable biobased materials as a substitute for petroleum-based feed stocks in the preparation of different types of industrially important polymers. Plant polyphenols, a kind of natural aromatic biomolecule, exist widely in some plant species. Benefiting from their special macromolecular structure, high reactivity, and broad abundance, plant polyphenols are potent candidates to replace the dwindling aromatic monomers derived from petroleum-based resources in synthesizing high-quality polymeric materials. In this review, the most related and innovative methods for elaborating novel polymeric materials from plant polyphenols are addressed. After a brief historical overview, the classification, structural characteristics, and reactivity of plant polyphenols are summarized in detail. In addition, some interesting and innovative works concerning the chemical modifications and polymerization techniques of plant polyphenols are also discussed. Importantly, the main chemical pathways to create plant polyphenol-based organic/organic-inorganic polymeric materials as well as their properties and possible applications are systematically described. We believe that this review could offer helpful references for designing multifunctional polyphenolic materials.
Collapse
Affiliation(s)
- Yang Liu
- Tianjin Fire Science and Technology Research Institute of MEM, Tianjin 300381, China;
- Key Laboratory of Fire Protection Technology for Industry and Public Building, Ministry of Emergency Management, Tianjin 300381, China
- Tianjin Key Laboratory of Fire Safety Technology, Tianjin 300381, China
| | - Junsheng Wang
- Tianjin Fire Science and Technology Research Institute of MEM, Tianjin 300381, China;
- Key Laboratory of Fire Protection Technology for Industry and Public Building, Ministry of Emergency Management, Tianjin 300381, China
- Tianjin Key Laboratory of Fire Safety Technology, Tianjin 300381, China
| | - Zhe Sun
- College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| |
Collapse
|
12
|
He Z, Liu Z, Chen Y. Chemical Design Strategy of Ionizable Lipids for In Vivo mRNA Delivery. ChemMedChem 2024; 19:e202400199. [PMID: 38722488 DOI: 10.1002/cmdc.202400199] [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: 03/17/2024] [Revised: 05/08/2024] [Indexed: 06/27/2024]
Abstract
Lipid nanoparticles (LNPs) are the most clinically successful drug delivery systems that have accelerated the development of mRNA drugs and vaccines. Among various structural components of LNPs, more recent attention has been paid in ionizable lipids (ILs) that was supposed as the key component in determining the effectiveness of LNPs for in vivo mRNA delivery. ILs are typically comprised of three moieties including ionizable heads, linkers, and hydrophobic tails, which suggested that the combination of different functional groups in three moieties could produce ILs with diverse chemical structures and biological identities. In this concept article, we provide a summary of chemical design strategy for high-performing IL candidates and discuss their structure-activity relationships for shifting tissue-selective mRNA delivery. We also propose an outlook for the development of next-generation ILs, enabling the broader translation of mRNA formulated with LNPs.
Collapse
Affiliation(s)
- Zepeng He
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhijia Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Functional Biomaterials Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510006, China
- College of Chemistry and Molecular Science, Henan University, Zhengzhou, 450046, China
- State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou, 450046, China
| |
Collapse
|
13
|
Liu J, Zhou Y, Lyu Q, Yao X, Wang W. Targeted protein delivery based on stimuli-triggered nanomedicine. EXPLORATION (BEIJING, CHINA) 2024; 4:20230025. [PMID: 38939867 PMCID: PMC11189579 DOI: 10.1002/exp.20230025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/07/2023] [Indexed: 06/29/2024]
Abstract
Protein-based drugs have shown unique advantages to treat various diseases in recent years. However, most protein therapeutics in clinical use are limited to extracellular targets with low delivery efficiency. To realize targeted protein delivery, a series of stimuli-triggered nanoparticle formulations have been developed to improve delivery efficiency and reduce off-target release. These smart nanoparticles are designed to release cargo proteins in response to either internal or external stimuli at pathological tissues. In this way, varieties of protein-based drugs including antibodies, enzymes, and pro-apoptotic proteins can be effectively delivered to desired sites for the treatment of cancer, inflammation, metabolic diseases, and so on with minimal side effects. In this review, recent advances in the design of stimuli-triggered nanomedicine for targeted protein delivery in different biomedical applications will be discussed. A deeper understanding of these emerging strategies helps develop more efficient protein delivery systems for clinical use in the future.
Collapse
Affiliation(s)
- Jinzhao Liu
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Yang Zhou
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Qingyang Lyu
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| | - Xiaotong Yao
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Department of ChemistryFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Weiping Wang
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong KongChina
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong KongChina
| |
Collapse
|
14
|
Ma Y, Tiwade PB, VanKeulen-Miller R, Narasipura EA, Fenton OS. Polyphenolic Nanoparticle Platforms (PARCELs) for In Vitro and In Vivo mRNA Delivery. NANO LETTERS 2024; 24:6092-6101. [PMID: 38728297 PMCID: PMC11218425 DOI: 10.1021/acs.nanolett.4c01235] [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] [Indexed: 05/12/2024]
Abstract
Despite their successful implementation in the COVID-19 vaccines, lipid nanoparticles (LNPs) still face a central limitation in the delivery of mRNA payloads: endosomal trapping. Improving upon this inefficiency could afford improved drug delivery systems, paving the way toward safer and more effective mRNA-based medicines. Here, we present polyphenolic nanoparticle platforms (PARCELs) as effective mRNA delivery systems. In brief, our investigation begins with a computationally guided structural analysis of 1825 discrete polyphenolic structural data points across 73 diverse small molecule polyphenols and 25 molecular parameters. We then generate structurally diverse PARCELs, evaluating their in vitro mechanism and activity, ultimately highlighting the superior endosomal escape properties of PARCELs relative to analogous LNPs. Finally, we examine the in vivo biodistribution, protein expression, and therapeutic efficacy of PARCELs in mice. In undertaking this approach, the goal of this study is to establish PARCELs as viable delivery platforms for safe and effective mRNA delivery.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Eshan Amruth Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen Shea Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
15
|
Song J, Fransen PPKH, Bakker MH, Wijnands SPW, Huang J, Guo S, Dankers PYW. The effect of charge and albumin on cellular uptake of supramolecular polymer nanostructures. J Mater Chem B 2024; 12:4854-4866. [PMID: 38682307 PMCID: PMC11111113 DOI: 10.1039/d3tb02631k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/30/2024] [Indexed: 05/01/2024]
Abstract
Intracellular delivery of functional biomolecules by using supramolecular polymer nanostructures has gained significant interest. Here, various charged supramolecular ureido-pyrimidinone (UPy)-aggregates were designed and formulated via a simple "mix-and-match" method. The cellular internalization of these UPy-aggregates in the presence or absence of serum proteins by phagocytic and non-phagocytic cells, i.e., THP-1 derived macrophages and immortalized human kidney cells (HK-2 cells), was systematically investigated. In the presence of serum proteins the UPy-aggregates were taken up by both types of cells irrespective of the charge properties of the UPy-aggregates, and the UPy-aggregates co-localized with mitochondria of the cells. In the absence of serum proteins only cationic UPy-aggregates could be effectively internalized by THP-1 derived macrophages, and the internalized UPy-aggregates either co-localized with mitochondria or displayed as vesicular structures. While the cationic UPy-aggregates were hardly internalized by HK-2 cells and could only bind to the membrane of HK-2 cells. With adding and increasing the amount of serum albumin in the cell culture medium, the cationic UPy-aggregates were gradually taken up by HK-2 cells without anchoring on the cell membranes. It is proposed that the serum albumin regulates the cellular internalization of UPy-aggregates. These results provide fundamental insights for the fabrication of supramolecular polymer nanostructures for intracellular delivery of therapeutics.
Collapse
Affiliation(s)
- Jiankang Song
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Peter-Paul K H Fransen
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Maarten H Bakker
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Sjors P W Wijnands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Jingyi Huang
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| | - Shuaiqi Guo
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands.
- Department of Biomedical Engineering, Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB, The Netherlands
| |
Collapse
|
16
|
Yin Y, Zhang Y, Xie Q, He Y, Guo J. Controlled Self-Assembly of Natural Polyphenols Driven by Multiple Molecular Interactions. Chempluschem 2024; 89:e202300695. [PMID: 38251920 DOI: 10.1002/cplu.202300695] [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: 11/28/2023] [Revised: 12/28/2023] [Indexed: 01/23/2024]
Abstract
Nature has exhibited a high degree of control over the structures and functions. Supramolecules have been utilized to mimic the subtle assembly in nature. However, sophisticated synthesis of molecular skeletons or programmable design of the driving forces raises great challenges in fabricating high-level superstructures in a controlled manner. Natural polyphenols show great promises as building blocks for a diverse of assemblies with controlled structures and functionalities. The intrinsically embedded phenolic groups (i. e., catechol and galloyl groups) are readily forming multiple molecular interactions, including coordination, hydrogen bonding, and π-π interactions with various materials of inorganic particles, organic compounds, synthetic polymers, and biomacromolecules, providing the self-assembled structures or nanocoating on surfaces. Subsequent assembly occurred by further bonding of polyphenols to construct supraparticles. To gain control over the self-assembly, the key lies in the interplay among the molecular interactions with one or two being dominant. In this Perspective, we introduce the representative polyphenol-based assemblies and their derived supraparticles to exhibit the effective harness of the controlled self-assembly by polyphenols.
Collapse
Affiliation(s)
- Yun Yin
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yajing Zhang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Qiuping Xie
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, Sichuan 610065, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, Sichuan 610065, China
- Bioproducts Institute, Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
17
|
Sarkar A, Sarkhel S, Bisht D, Jaiswal A. Cationic dextrin nanoparticles for effective intracellular delivery of cytochrome C in cancer therapy. RSC Chem Biol 2024; 5:249-261. [PMID: 38456040 PMCID: PMC10915965 DOI: 10.1039/d3cb00090g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/19/2023] [Indexed: 03/09/2024] Open
Abstract
Intracellular protein delivery shows promise as a selective and specific approach to cancer therapy. However, a major challenge is posed by delivering proteins into the target cells. Despite the development of nanoparticle (NP)-based approaches, a versatile and biocompatible delivery system that can deliver active therapeutic cargo into the cytosol while escaping endosome degradation remains elusive. In order to overcome these challenges, a polymeric nanocarrier was prepared using cationic dextrin (CD), a biocompatible and biodegradable polymer, to encapsulate and deliver cytochrome C (Cyt C), a therapeutic protein. The challenge of endosomal escape of the nanoparticles was addressed by co-delivering the synthesized NP construct with chloroquine, which enhances the endosomal escape of the therapeutic protein. No toxicity was observed for both CD NPs and chloroquine at the concentration tested in this study. Spectroscopic investigations confirmed that the delivered protein, Cyt C, was structurally and functionally active. Additionally, the delivered Cyt C was able to induce apoptosis by causing depolarization of the mitochondrial membrane in HeLa cells, as evidenced by flow cytometry and microscopic observations. Our findings demonstrate that an engineered delivery system using CD NPs is a promising platform in nanomedicine for protein delivery applications.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Sanchita Sarkhel
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Deepali Bisht
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| | - Amit Jaiswal
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi Kamand Mandi 175075 Himachal Pradesh India
| |
Collapse
|
18
|
Li S, Ma Y, Cui J, Caruso F, Ju Y. Engineering poly(ethylene glycol) particles for targeted drug delivery. Chem Commun (Camb) 2024; 60:2591-2604. [PMID: 38285062 DOI: 10.1039/d3cc06098e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Poly(ethylene glycol) (PEG) is considered to be the "gold standard" among the stealth polymers employed for drug delivery. Using PEG to modify or engineer particles has thus gained increasing interest because of the ability to prolong blood circulation time and reduce nonspecific biodistribution of particles in vivo, owing to the low fouling and stealth properties of PEG. In addition, endowing PEG-based particles with targeting and drug-loading properties is essential to achieve enhanced drug accumulation at target sites in vivo. In this feature article, we focus on recent work on the synthesis of PEG particles, in which PEG is the main component in the particles. We highlight different synthesis methods used to generate PEG particles, the influence of the physiochemical properties of PEG particles on their stealth and targeting properties, and the application of PEG particles in targeted drug delivery.
Collapse
Affiliation(s)
- Shiyao Li
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia.
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yi Ju
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia.
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
19
|
Guo Y, Zhai X, Li N, Zan X. Recent Progress in Protein-Polyphenol Assemblies for Biomedical Applications. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2005-2014. [PMID: 38227800 DOI: 10.1021/acs.langmuir.3c03244] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Nowadays, natural materials as smart building blocks for assembling functional materials have aroused extensive interest in the scientific community. Proteins and polyphenols are typical natural building blocks that are widely used. On the one hand, proteins are one of the most versatile classes of biomolecules, serving as catalysts, signaling molecules, transporters, receptors, scaffolds that maintain the integrity of cell and tissue, and more. On the other hand, the facile adhesion of naturally abundant polyphenols with other substances and their potential biomedical applications have been highly attractive for functional biomaterials fabrication. Additionally, there are a variety of interactions between the proteins and polyphenols, mainly hydrogen bonding, hydrophobic, and ionic interactions. These reversible dynamic interactions enable proteins and polyphenols to form stable protein-polyphenol assemblies and maintain their inherent structures and biological activities in the assemblies. Therefore, protein-polyphenol assemblies can be applied to design a variety of advanced functional materials for biomedical applications. Herein, recent progress in protein-polyphenol particles, capsules, coatings, and hydrogels is summarized, the preparation and application of these assemblies are introduced in detail, and the future of the field is prospected.
Collapse
Affiliation(s)
- Yan Guo
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering, Hunan University of Science and Technology, Hunan 411201, China
| | - Xinyue Zhai
- Hunan Provincial Key Laboratory of Advanced Materials for New Energy Storage and Conversion, School of Materials Science and Engineering, Hunan University of Science and Technology, Hunan 411201, China
| | - Na Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou 325001, China
| | - Xingjie Zan
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou 325001, China
| |
Collapse
|
20
|
Lv Y, Li W, Liao W, Jiang H, Liu Y, Cao J, Lu W, Feng Y. Nano-Drug Delivery Systems Based on Natural Products. Int J Nanomedicine 2024; 19:541-569. [PMID: 38260243 PMCID: PMC10802180 DOI: 10.2147/ijn.s443692] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Natural products have proven to have significant curative effects and are increasingly considered as potential candidates for clinical prevention, diagnosis, and treatment. Compared with synthetic drugs, natural products not only have diverse structures but also exhibit a range of biological activities against different disease states and molecular targets, making them attractive for development in the field of medicine. Despite advancements in the use of natural products for clinical purposes, there remain obstacles that hinder their full potential. These challenges include issues such as limited solubility and stability when administered orally, as well as short durations of effectiveness. To address these concerns, nano-drug delivery systems have emerged as a promising solution to overcome the barriers faced in the clinical application of natural products. These systems offer notable advantages, such as a large specific surface area, enhanced targeting capabilities, and the ability to achieve sustained and controlled release. Extensive in vitro and in vivo studies have provided further evidence supporting the efficacy and safety of nanoparticle-based systems in delivering natural products in preclinical disease models. This review describes the limitations of natural product applications and the current status of natural products combined with nanotechnology. The latest advances in nano-drug delivery systems for delivery of natural products are considered from three aspects: connecting targeting warheads, self-assembly, and co-delivery. Finally, the challenges faced in the clinical translation of nano-drugs are discussed.
Collapse
Affiliation(s)
- Ying Lv
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Wenqing Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Wei Liao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Haibo Jiang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Yuwei Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Jiansheng Cao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Wenfei Lu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| | - Yufei Feng
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, 150040, People’s Republic of China
| |
Collapse
|
21
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Ma Y, Zhu G, Feng L, Jiang S, Xiang Q, Wang J. Efficient Cytotoxicity of Recombinant Azurin in Escherichia coli Nissle 1917-Derived Minicells against Colon Cancer Cells. Bioengineering (Basel) 2023; 10:1188. [PMID: 37892918 PMCID: PMC10603951 DOI: 10.3390/bioengineering10101188] [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: 05/26/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 10/29/2023] Open
Abstract
Compared to chemical drugs, therapeutic proteins exhibit higher specificity and activity and are generally well-tolerated by the human body. However, the limitations, such as poor stability both in vivo and in vitro as well as difficulties in penetrating cell membranes, hinder their widespread application. To overcome the challenges, a highly efficient protocol was developed and implemented for the recombinant expression of the therapeutic protein azurin and secretion into minicells derived from probiotic Escherichia coli Nissle 1917. The novel coupled production with a delivery system of therapeutic proteins based on minicells was obtained through purification to enhance protein activity, circulation characteristics, and targeting specificity. This protein drug carrier integrates the production of carrier materials and the loading of expression proteins. The drug carrier also protects the encapsulated polypeptide drugs from enzymatic or gastric acid degradation until they are released. Escherichia coli Nissle 1917-derived minicells have natural targeting to colon cancer cells, low toxicity, and can accumulate for a long time after penetrating tumor tissue. This self-produced protein drug delivery system simplified the process of protein preparation, and its inhibitory effect on different types of colon cancer cells was verified by CCK-8 cytotoxicity assay, cancer cell invasion, and migration assay. This work provided a simple method to prepare minicell drug delivery systems for protein drug production and a novel approach for the transport of recombinant protein drugs.
Collapse
Affiliation(s)
- Yi Ma
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Guanshu Zhu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lan Feng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Shoujin Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Jufang Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
23
|
Ma Y, Fenton OS. A Unified Strategy to Improve Lipid Nanoparticle Mediated mRNA Delivery Using Adenosine Triphosphate. J Am Chem Soc 2023; 145:19800-19811. [PMID: 37656876 DOI: 10.1021/jacs.3c05574] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023]
Abstract
A central goal of chemical and drug delivery sciences is to maximize the therapeutic efficacy of a given drug at the lowest possible dose. Here, we report a generalizable strategy that can be utilized to improve the delivery of mRNA drugs using lipid nanoparticles (LNPs), the clinically approved chemistry platforms utilized in the Moderna and Pfizer/BioNTech COVID-19 vaccines. In brief, our strategy updates the chemistry of LNPs to incorporate adenosine triphosphate (ATP) alongside mRNA, a modification that results in upward of a 79-fold increase in LNP-delivered mRNA-encoded protein expression in vitro and a 24-fold increase in vivo when compared to parent mRNA LNP formulations that do not contain ATP. Notably, we find that our ATP co-delivery strategy increases LNP-delivered mRNA-encoded protein expression across eight different LNP chemistries and three different cell lines, under normoxia and hypoxia, and in a well-tolerated fashion. Notably, our strategy also improves the expression of mRNA encoding for intracellular and secreted proteins both in vitro and in vivo, highlighting the utility of leveraging ATP co-delivery within mRNA LNPs as a means to increase protein expression. In developing this strategy, we hope that we have provided a simple yet powerful approach to improving mRNA LNPs that may one day be useful in developing therapies for human disease.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
24
|
Wang N, Zhang G, Zhang P, Zhao K, Tian Y, Cui J. Vaccination of TLR7/8 Agonist-Conjugated Antigen Nanoparticles for Cancer Immunotherapy. Adv Healthc Mater 2023; 12:e2300249. [PMID: 37016572 DOI: 10.1002/adhm.202300249] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/14/2023] [Indexed: 04/06/2023]
Abstract
Nanovaccine-based immunotherapy can initiate strong immune responses and establish a long-term immune memory to prevent tumor invasion and recurrence. Herein, the assembly of redox-responsive antigen nanoparticles (NPs) conjugated with imidazoquinoline-based TLR7/8 agonists for lymph node-targeted immune activation is reported, which can potentiate tumor therapy and prevention. Antigen NPs are assembled via the templating of zeolitic imidazolate framework-8 NPs to cross-link ovalbumin with disulfide bonds, which enables the NPs with redox-responsiveness for improved antigen cross-presentation and dendritic cell activation. The formulated nanovaccines promote the lymphatic co-delivery of antigens and agonists, which can trigger immune responses of cytotoxic T lymphocytes and strong immunological memory. Notably, nanovaccines demonstrate their superiority for tumor prevention owing to the elicited robust antitumor immunity. The reported strategy provides a rational design of nanovaccines for enhanced cancer immunotherapy.
Collapse
Affiliation(s)
- Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Guiqiang Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Peiyu Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Kaijie Zhao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Yuan Tian
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, P. R. China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, P. R. China
| |
Collapse
|
25
|
Ma H, Xing F, Zhou Y, Yu P, Luo R, Xu J, Xiang Z, Rommens PM, Duan X, Ritz U. Design and fabrication of intracellular therapeutic cargo delivery systems based on nanomaterials: current status and future perspectives. J Mater Chem B 2023; 11:7873-7912. [PMID: 37551112 DOI: 10.1039/d3tb01008b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Intracellular cargo delivery, the introduction of small molecules, proteins, and nucleic acids into a specific targeted site in a biological system, is an important strategy for deciphering cell function, directing cell fate, and reprogramming cell behavior. With the advancement of nanotechnology, many researchers use nanoparticles (NPs) to break through biological barriers to achieving efficient targeted delivery in biological systems, bringing a new way to realize efficient targeted drug delivery in biological systems. With a similar size to many biomolecules, NPs possess excellent physical and chemical properties and a certain targeting ability after functional modification on the surface of NPs. Currently, intracellular cargo delivery based on NPs has emerged as an important strategy for genome editing regimens and cell therapy. Although researchers can successfully deliver NPs into biological systems, many of them are delivered very inefficiently and are not specifically targeted. Hence, the development of efficient, target-capable, and safe nanoscale drug delivery systems to deliver therapeutic substances to cells or organs is a major challenge today. In this review, on the basis of describing the research overview and classification of NPs, we focused on the current research status of intracellular cargo delivery based on NPs in biological systems, and discuss the current problems and challenges in the delivery process of NPs in biological systems.
Collapse
Affiliation(s)
- Hong Ma
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Fei Xing
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Ludwigstraße 23, 35392 Giessen, Germany
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rong Luo
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiawei Xu
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedic Surgery, The Fifth People's Hospital of Sichuan Province, Chengdu, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
26
|
Yu Q, Zhou J, Wang H, Liu Y, Zhou H, Kang B, Chen HY, Xu JJ. A Multiple-Response Cascade Nanoreactor for Starvation and Deep Catalysis Chemodynamic Assisted Near-Infrared-II Mild Photothermal Therapy. CHEMICAL & BIOMEDICAL IMAGING 2023; 1:242-250. [PMID: 39473696 PMCID: PMC11503788 DOI: 10.1021/cbmi.2c00003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 11/29/2024]
Abstract
Photothermal therapy (PTT) is hampered by the limited capacity of light penetration, non-specific thermal diffusion damage to surrounding healthy tissues, and thermoresistance originating in heat shock proteins (HSPs). Here, a triple-respondent (triphosphate, glutathione, and pH) cascade nanoreactor was designed through the glucose consumption-induced thermal sensitization strategy. The near-infrared-II (NIR-II) photothermal reagent Bi-Au was encapsulated in a glucose oxidase-based protein-polyphenol structure with the help of the zeolitic imidazolate framework-8 (ZIF-8). The composite nanosystem possesses triple-enzyme activity (glucose oxidase, peroxidase-like, and catalase-like). On one hand, the product of hydrogen peroxide from glycolysis can be converted into reactive oxygen species and oxygen to enhance the chemodynamic therapy and alleviate the state of hypoxia in tumor cells. On the other hand, glucose consumption could down-regulate the expression level of HSPs. The heat resistance ability of cells decreased under starvation and oxidative damage states, which is beneficial to reduce the temperature required for PTT and improve the efficiency of mild PTT. The cascade nanocapsule exhibited high tumor inhibition and proposed a typical synergistic strategy for starvation, chemodynamic, and NIR-II mild photothermal therapy.
Collapse
Affiliation(s)
- Qiao Yu
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Jie Zhou
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Hui Wang
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Yong Liu
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Hong Zhou
- Key
Laboratory of Optic-electric Sensing and Analytical Chemistry for
Life Science, Ministry of Education, College of Chemistry and Molecular
Engineering, Qingdao University of Science
and Technology, Qingdao266042, P.R. China
| | - Bin Kang
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Hong-Yuan Chen
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| | - Jing-Juan Xu
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing210023, P.R. China
| |
Collapse
|
27
|
Ma Y, Fenton OS. An Efficacy and Mechanism Driven Study on the Impact of Hypoxia on Lipid Nanoparticle Mediated mRNA Delivery. J Am Chem Soc 2023; 145:11375-11386. [PMID: 37184377 DOI: 10.1021/jacs.3c02584] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Hypoxia is a common hallmark of human disease that is characterized by abnormally low oxygen levels in the body. While the effects of hypoxia on many small molecule-based drugs are known, its effects on several classes of next-generation medications including messenger RNA therapies warrant further study. Here, we provide an efficacy- and mechanism-driven study that details how hypoxia impacts the cellular response to mRNA therapies delivered using 4 different chemistries of lipid nanoparticles (LNPs, the frontrunner class of drug delivery vehicles for translational mRNA therapy utilized in the Moderna and Pfizer/BioNTech COVID-19 vaccines). Specifically, our work provides a comparative analysis as to how various states of oxygenation impact LNP-delivered mRNA expression, cellular association, endosomal escape, and intracellular ATP concentrations following treatment with 4 different LNPs across 3 different cell lines. In brief, we first identify that hypoxic cells express less LNP-delivered mRNA into protein than normoxic cells. Next, we identify generalizable cellular reoxygenation protocols that can reverse the negative effects that hypoxia imparts on LNP-delivered mRNA expression. Finally, mechanistic studies that utilize fluorescence-activated cell sorting, confocal microscopy, and enzyme inhibition reveal that decreases in mRNA expression correlate with decreases in intracellular ATP (rather than with differences in mRNA LNP uptake pathways). In presenting this data, we hope that our work provides a comprehensive efficacy and mechanism-driven study that explores the impact of differential oxygenation on LNP-delivered mRNA expression while simultaneously establishing fundamental criteria that may one day be useful for the development of mRNA drugs to treat hypoxia-associated disease.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
28
|
Kim KH, Ki MR, Min KH, Pack SP. Advanced Delivery System of Polyphenols for Effective Cancer Prevention and Therapy. Antioxidants (Basel) 2023; 12:antiox12051048. [PMID: 37237914 DOI: 10.3390/antiox12051048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Polyphenols from plants such as fruits and vegetables are phytochemicals with physiological and pharmacological activity as potential drugs to modulate oxidative stress and inflammation associated with cardiovascular disease, chronic disease, and cancer. However, due to the limited water solubility and bioavailability of many natural compounds, their pharmacological applications have been limited. Researchers have made progress in the development of nano- and micro-carriers that can address these issues and facilitate effective drug delivery. The currently developed drug delivery systems maximize the fundamental effects in various aspects such as absorption rate, stability, cellular absorption, and bioactivity of polyphenols. This review focuses on the antioxidant and anti-inflammatory effects of polyphenols enhanced by the introduction of drug delivery systems, and ultimately discusses the inhibition of cancer cell proliferation, growth, and angiogenesis.
Collapse
Affiliation(s)
- Koung Hee Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea
| | - Mi-Ran Ki
- Institute of Industrial Technology, Korea University, Sejong 30019, Republic of Korea
| | - Ki Ha Min
- Institute of Industrial Technology, Korea University, Sejong 30019, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
29
|
Zhang J, Gao B, Ye B, Sun Z, Qian Z, Yu L, Bi Y, Ma L, Ding Y, Du Y, Wang W, Mao Z. Mitochondrial-Targeted Delivery of Polyphenol-Mediated Antioxidases Complexes against Pyroptosis and Inflammatory Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208571. [PMID: 36648306 DOI: 10.1002/adma.202208571] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/30/2022] [Indexed: 06/17/2023]
Abstract
Excess accumulation of mitochondrial reactive oxygen species (mtROS) is a key target for inhibiting pyroptosis-induced inflammation and tissue damage. However, targeted delivery of drugs to mitochondria and efficient clearance of mtROS remain challenging. In current study, it is discovered that polyphenols such as tannic acid (TA) can mediate the targeting of polyphenol/antioxidases complexes to mitochondria. This affinity does not depend on mitochondrial membrane potential but stems from the strong binding of TA to mitochondrial outer membrane proteins. Taking advantage of the feasibility of self-assembly between TA and proteins, superoxide dismutase, catalase, and TA are assembled into complexes (referred to as TSC) for efficient enzymatic activity maintenance. In vitro fluorescence confocal imaging shows that TSC not only promoted the uptake of biological enzymes in hepatocytes but also highly overlapped with mitochondria after lysosomal escape. The results from an in vitro model of hepatocyte oxidative stress demonstrate that TSC efficiently scavenges excess mtROS and reverses mitochondrial depolarization, thereby inhibiting inflammasome-mediated pyroptosis. More interestingly, TSC maintain superior efficacy compared with the clinical gold standard drug N-acetylcysteine in both acetaminophen- and D-galactosamine/lipopolysaccharide-induced pyroptosis-related hepatitis mouse models. In conclusion, this study opens a new paradigm for targeting mitochondrial oxidative stress to inhibit pyroptosis and treat inflammatory diseases.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Bingqiang Gao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Binglin Ye
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Lisha Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yanli Bi
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yang Du
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, 310009, P. R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, 310058, P. R. China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, P. R. China
| |
Collapse
|
30
|
Fan J, Yu H, Lu X, Xue R, Guan J, Xu Y, Qi Y, He L, Yu W, Abay S, Li Z, Huo S, Li L, Lv M, Li W, Chen W, Han B. Overlooked Spherical Nanoparticles Exist in Plant Extracts: From Mechanism to Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8854-8871. [PMID: 36757908 DOI: 10.1021/acsami.2c19065] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
To date, plant medicine research has focused mainly on the chemical compositions of plant extracts and their medicinal effects. However, the therapeutic or toxic effects of nanoparticles in plant extracts remain unclear. In this study, large numbers of spherical nanoparticles were discovered in some plant extracts. Nanoparticles in Turkish galls extracts were used as an example to examine their pH responsiveness, free radical scavenging, and antibacterial capabilities. By utilizing the underlying formation mechanism of these nanoparticles, a general platform to produce spherical nanoparticles via direct self-assembly of Turkish gall extracts and various functional proteins was developed. The results showed that the nanoparticles retained both the antibacterial ability and intracellular carrier ability of the original protein or catechol. This work introduces a new member of the plant-derived edible nanoparticle (PDEN) family, establishes a simple and versatile platform for mass production nanoparticles, and provides new insight into the formation mechanism of nanoparticles during plant extraction.
Collapse
Affiliation(s)
- Jingmin Fan
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Hang Yu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Xin Lu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Rui Xue
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Jiawei Guan
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Yu Xu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Yunyun Qi
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Linyun He
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Wei Yu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Sirapil Abay
- Xinjiang Institute of Traditional Uygur Medicine, Urumqi 830049, China
| | - Zhijian Li
- Xinjiang Institute of Traditional Uygur Medicine, Urumqi 830049, China
| | - Shixia Huo
- Xinjiang Institute of Traditional Uygur Medicine, Urumqi 830049, China
| | - Le Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Mengying Lv
- Department of Pharmacy/The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Wenxin Li
- Laboratory of Nano-biology and Medicine, Shanghai Institute of Applied Physics, Shanghai 201800, China
| | - Wen Chen
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| | - Bo Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi University, Shihezi 832002, China
| |
Collapse
|
31
|
Wang Y, Chen X, Xu X, Du M, Wu C. Reducing disulfide bonds as a robust strategy to facilitate the self-assembly of cod protein fabricating potential active ingredients-nanocarrier. Colloids Surf B Biointerfaces 2023; 222:113080. [PMID: 36542952 DOI: 10.1016/j.colsurfb.2022.113080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/24/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
In this study, a novel method was developed to encapsulate hydrophobic compounds by self-assembly of cod protein (CP) triggered by breaking disulfide bonds. Curcumin (Cur), a representative lipid-soluble polyphenol, was selected as a model to evaluate the potential of CP nanoparticles as novel and accessible nanocarriers. Results showed that the protein structure gradually unfolded with increasing dithiothreitol (DTT) concentration, indicating that S-S cleavage was conducive to forming a looser structure. The resultant unfolded CP exposed more hydrophobic sites, facilitating its interaction with hydrophobic compounds. The encapsulation efficiency (EE) of formed CP-Cur nanoparticles was relatively high, reaching 99.09%, 98.8%, and 89.77% when the mass ratios of CP to Cur were 20:1, 10:1, and 5:1 (w/v), respectively. The hydrophobic interaction, weak van der Waals, and hydrogen bond were the forces contributing to the formation of CP-Cur nanoparticles, whereas the hydrophobic interaction played a crucial role. The CP-Cur complex exhibited increased stability and a homogeneous-stable structural phase. Thus, this research not only proposed a novel and simple encapsulation method of hydrophobic bioactive compounds but also provided a theoretical reference for the application of reductants in food or pharmacy system.
Collapse
Affiliation(s)
- Yuying Wang
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China; College of Food Science, Jilin University, Changchun 130015, China
| | - Xufei Chen
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Xianbing Xu
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Ming Du
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China
| | - Chao Wu
- National Engineering Research Center of Seafood, Dalian 116034, China; College of Food Science, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
32
|
Tong MQ, Lu CT, Huang LT, Yang JJ, Yang ST, Chen HB, Xue PP, Luo LZ, Yao Q, Xu HL, Zhao YZ. Polyphenol-driven facile assembly of a nanosized acid fibroblast growth factor-containing coacervate accelerates the healing of diabetic wounds. Acta Biomater 2023; 157:467-486. [PMID: 36460288 DOI: 10.1016/j.actbio.2022.11.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022]
Abstract
Diabetic wounds are challenging to heal due to complex pathogenic abnormalities. Routine treatment with acid fibroblast growth factor (aFGF) is widely used for diabetic wounds but hardly offers a satisfying outcome due to its instability. Despite the emergence of various nanoparticle-based protein delivery approaches, it remains challenging to engineer a versatile delivery system capable of enhancing protein stability without the need for complex preparation. Herein, a polyphenol-driven facile assembly of nanosized coacervates (AE-NPs) composed of aFGF and Epigallocatechin-3-gallate (EGCG) was constructed and applied in the healing of diabetic wounds. First, the binding patterns of EGCG and aFGF were predicted by molecular docking analysis. Then, the characterizations demonstrated that AE-NPs displayed higher stability in hostile conditions than free aFGF by enhancing the binding activity of aFGF to cell surface receptors. Meanwhile, the AE-NPs also had a powerful ability to scavenge reactive oxygen species (ROS) and promote angiogenesis, which significantly accelerated full-thickness excisional wound healing in diabetic mice. Besides, the AE-NPs suppressed the early scar formation by improving collagen remodeling and the mechanism was associated with the TGF-β/Smad signaling pathway. Conclusively, AE-NPs might be a potential and facile strategy for stabilizing protein drugs and achieving the scar-free healing of diabetic wounds. STATEMENT OF SIGNIFICANCE: Diabetic chronic wound is among the serious complications of diabetes that eventually cause the amputation of limbs. Herein, a polyphenol-driven facile assembly of nanosized coacervates (AE-NPs) composed of aFGF and EGCG was constructed. The EGCG not only acted as a carrier but also possessed a therapeutic effect of ROS scavenging. The AE-NPs enhanced the binding activity of aFGF to cell surface receptors on the cell surface, which improved the stability of aFGF in hostile conditions. Moreover, AE-NPs significantly accelerated wound healing and improved collagen remodeling by regulating the TGF-β/Smad signaling pathway. Our results bring new insights into the field of polyphenol-containing nanoparticles, showing their potential as drug delivery systems of macromolecules to treat diabetic wounds.
Collapse
Affiliation(s)
- Meng-Qi Tong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Cui-Tao Lu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lan-Tian Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiao-Jiao Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Si-Ting Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hang-Bo Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peng-Peng Xue
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lan-Zi Luo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qing Yao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - He-Lin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang 325000, China.
| | - Ying-Zheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang 325000, China.
| |
Collapse
|
33
|
Yu Q, Zhou J, Song J, Zhou H, Kang B, Chen HY, Xu JJ. A Cascade Nanoreactor of Metal-Protein-Polyphenol Capsule for Oxygen-Mediated Synergistic Tumor Starvation and Chemodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206592. [PMID: 36437115 DOI: 10.1002/smll.202206592] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/05/2022] [Indexed: 06/16/2023]
Abstract
Starvation therapy kills tumor cells via consuming glucose to cut off their energy supply. However, since glucose oxidase (GOx)-mediated glycolysis is oxygen-dependent, the cascade reaction based on GOx faces the challenge of a hypoxic tumor microenvironment. By decomposition of glycolysis production of H2 O2 into O2 , starvation therapy can be enhanced, but chemodynamic therapy is limited. Here, a close-loop strategy for on demand H2 O2 and O2 delivery, release, and recycling is proposed. The nanoreactor (metal-protein-polyphenol capsule) is designed by incorporating two native proteins, GOx and hemoglobin (Hb), in polyphenol networks with zeolitic imidazolate framework as sacrificial templates. Glycolysis occurs in the presence of GOx with O2 consumption and the produced H2 O2 reacts with Hb to produce highly cytotoxic hydroxyl radicals (•OH) and methemoglobin (MHb) (Fenton reaction). Benefiting from the different oxygen carrying capacities of Hb and MHb, oxygen on Hb is rapidly released to supplement its consumption during glycolysis. Glycolysis and Fenton reactions are mutually reinforced by oxygen supply, consuming more glucose and producing more hydroxyl radicals and ultimately enhancing both starvation therapy and chemodynamic therapy. This cascade nanoreactor exhibits high efficiency for tumor suppression and provides an effective strategy for oxygen-mediated synergistic starvation therapy and chemodynamic therapy.
Collapse
Affiliation(s)
- Qiao Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Jie Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Juan Song
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Hong Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, P. R. China
| | - Bin Kang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
34
|
Zhou T, Yuan S, Qian P, Wu Y. Enzymes in Nanomedicine for Anti-tumor Therapy. Chem Res Chin Univ 2023. [DOI: 10.1007/s40242-023-2349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
35
|
Yin Z, Zhang Z, Gao D, Luo G, Ma T, Wang Y, Lu L, Gao X. Stepwise Coordination-Driven Metal-Phenolic Nanoparticle as a Neuroprotection Enhancer for Alzheimer's Disease Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:524-540. [PMID: 36542560 DOI: 10.1021/acsami.2c18060] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Current therapeutic strategies for Alzheimer's disease (AD) mainly focus on inhibition of aberrant amyloid-β peptide (Aβ) aggregation. However, these strategies cannot repair the side symptoms (e.g., high neuronal oxidative stress) triggered by Aβ accumulation and thus show limited effects on suppressing Aβ-induced neuronal apoptosis. Herein, we develop a stepwise metal-phenolic coordination approach for the rational design of a neuroprotection enhancer, K8@Fe-Rh/Pda NPs, in which rhein and polydopamine are effectively coupled to enhance the treatment of AD in APPswe/PSEN1dE9 transgenic (APP/PS1) mice. We discover that the polydopamine inhibits the aggregation of Aβ oligomers, and rhein helps repair damage to neurons triggered by Aβ aggregation. Based on molecular docking, we demonstrate that the polydopamine has a strong interaction with Aβ monomers/fibrils through its multiple recognition sites (e.g., catechol groups, imine groups, and indolic/catecholic π-systems), thereby reducing Aβ burden. Further investigation of the antioxidant mechanisms suggests that K8@Fe-Rh/Pda NPs promote the mitochondrial biogenesis via activating the sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha pathway. This finally inhibits neuronal apoptosis. Moreover, an intravenous injection of these nanoparticles potently improves the cognitive function in APP/PS1 mice without adverse effects. Overall, our work provides a promising approach to develop advanced nanomaterials for multi-target treatment of AD.
Collapse
Affiliation(s)
- Zhihui Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| | - Zhixin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| | - Demin Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| | - Tao Ma
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing100078, China
| | - Ying Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| | - Lehui Lu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun130022, China
| | - Xiaoyan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing102488, China
| |
Collapse
|
36
|
A novel IONP-decorated two-dimensional [Zn2+]:[Insulin] nanosheet with ordered array of surface channels and cellular uptake potential. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Li N, Chen L, Luo Z, Nie G, Zhang P, He S, Peng J. Dual-Targeting of Doxorubicin and Chlorine e6 Co-Delivery Based on Small-Size Nanocomposite for the Synergetic Imaging and Therapy. J CLUST SCI 2022. [DOI: 10.1007/s10876-021-02098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Ju Y, Liao H, Richardson JJ, Guo J, Caruso F. Nanostructured particles assembled from natural building blocks for advanced therapies. Chem Soc Rev 2022; 51:4287-4336. [PMID: 35471996 DOI: 10.1039/d1cs00343g] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Advanced treatments based on immune system manipulation, gene transcription and regulation, specific organ and cell targeting, and/or photon energy conversion have emerged as promising therapeutic strategies against a range of challenging diseases. Naturally derived macromolecules (e.g., proteins, lipids, polysaccharides, and polyphenols) have increasingly found use as fundamental building blocks for nanostructured particles as their advantageous properties, including biocompatibility, biodegradability, inherent bioactivity, and diverse chemical properties make them suitable for advanced therapeutic applications. This review provides a timely and comprehensive summary of the use of a broad range of natural building blocks in the rapidly developing field of advanced therapeutics with insights specific to nanostructured particles. We focus on an up-to-date overview of the assembly of nanostructured particles using natural building blocks and summarize their key scientific and preclinical milestones for advanced therapies, including adoptive cell therapy, immunotherapy, gene therapy, active targeted drug delivery, photoacoustic therapy and imaging, photothermal therapy, and combinational therapy. A cross-comparison of the advantages and disadvantages of different natural building blocks are highlighted to elucidate the key design principles for such bio-derived nanoparticles toward improving their performance and adoption. Current challenges and future research directions are also discussed, which will accelerate our understanding of designing, engineering, and applying nanostructured particles for advanced therapies.
Collapse
Affiliation(s)
- Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia. .,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Haotian Liao
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Sichuan 610065, China
| | - Joseph J Richardson
- Department of Materials Engineering, University of Tokyo, 7-3-1 Bunkyo-ku, Tokyo 113-8656, Japan
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China. .,Bioproducts Institute, Departments of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
39
|
Rajendran K, Karthikeyan A, Krishnan UM. Emerging trends in nano-bioactive-mediated mitochondria-targeted therapeutic stratagems using polysaccharides, proteins and lipidic carriers. Int J Biol Macromol 2022; 208:627-641. [PMID: 35341885 DOI: 10.1016/j.ijbiomac.2022.03.121] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
Abstract
The emergence of new lifestyle disorders and pharmaco-resistant variants of diseases has necessitated the search for effective therapeutic moieties and approaches that could overcome the limitations in the existing treatment modalities. In this context, bioactives such as flavonoids, polyphenols, tannins, terpenoids and alkaloids have demonstrated promise in therapy owing to their ability to scavenge free radicals and modulate the mitochondrial function as well as regulate metabolic pathways. However, their clinical applicability is low owing to their poor bioavailability and aqueous solubility. The encapsulation of bioactives in nanodimensional particles has overcome these limitations to a large extent while simultaneously conferring additional advantages of improved circulation time, enhanced cell uptake and target specific release. A wide range of nanocarriers derived from biopolymers such as polysaccharides, lipids and proteins, have been explored for encapsulation of different bioactives and have reported significant improvement of the bioavailability and therapeutic efficacy of the encapsulated cargo. However, incorporation of cell-specific and mitochondria-specific elements on the nanocarriers has been relatively less explored. This review summarizes some of the recent attempts to treat different disorders using bioactives encapsulated in biopolymer nanostructures and few instances of mitochondria-specific delivery.
Collapse
Affiliation(s)
- Kayalvizhi Rajendran
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613 401, India; School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India
| | - Akhilasree Karthikeyan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613 401, India; School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613 401, India; School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur 613 401, India.
| |
Collapse
|
40
|
Jaiswal N, Halder S, Mahata N, Chanda N. Bi-Functional Gold Nanorod-Protein Conjugates with Biomimetic BSA@Folic Acid Corona for Improved Tumor Targeting and Intracellular Delivery of Therapeutic Proteins in Colon Cancer 3D Spheroids. ACS APPLIED BIO MATERIALS 2022; 5:1476-1488. [PMID: 35285613 DOI: 10.1021/acsabm.1c01216] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Gold nanorods (AuNRs) remain well-developed inorganic nanocarriers of small molecules for a plethora of biomedical and therapeutic applications. However, the delivery of therapeutic proteins using AuNRs with high protein loading capacity (LC), serum stability, excellent target specificity, and minimal off-target protein release is not known. Herein, we report two bi-functional AuNR-protein nanoconjugates, AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA, supramolecularly coated with folic acid-modified BSA (BSAFA) acting as biomimetic protein corona to demonstrate targeted cytosolic delivery of enhanced green fluorescent protein (EGFP) and therapeutic ribonuclease A enzyme (RNase A) in their functional forms. AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA exhibit high LCs of ∼42 and ∼54%, respectively, increased colloidal stability, and rapid protein release in the presence of biological thiols. As a nanocarrier, AuNR@EGFP-BSAFA and AuNR@RNaseA-BSAFA show resistance to corona formation in high-serum media even after 24 h, guaranteeing a greater circulation lifetime. Folate receptor-targeting BSAFA on the AuNR surface facilitates the receptor-mediated internalization, followed by the release of EGFP and RNase A in HT29 cells. The green fluorescence dispersed throughout the cell's cytoplasm indicates successful cytosolic delivery of EGFP by AuNR@EGFP-BSAFA. AuNR@RNaseA-BSAFA-mediated therapeutic RNase A delivery in multicellular 3D spheroids of HT29 cells exhibits a radical reduction in the cellular RNA fluorescence intensity to 38%, signifying RNA degradation and subsequent cell death. The versatile nanoformulation strategy in terms of the anisotropic particle morphology, protein type, and ability for targeted delivery in the functional form makes the present AuNR-protein nanoconjugates a promising platform for potential application in cancer management.
Collapse
Affiliation(s)
- Namita Jaiswal
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India.,Material Processing and Microsystem Laboratory, CSIR─Central Mechanical Engineering Research Institute, Durgapur 713209, India
| | - Sudeshna Halder
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nripen Chanda
- Material Processing and Microsystem Laboratory, CSIR─Central Mechanical Engineering Research Institute, Durgapur 713209, India
| |
Collapse
|
41
|
Chen J, Pan S, Zhou J, Lin Z, Qu Y, Glab A, Han Y, Richardson JJ, Caruso F. Assembly of Bioactive Nanoparticles via Metal-Phenolic Complexation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108624. [PMID: 34933398 DOI: 10.1002/adma.202108624] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The integration of bioactive materials (e.g., proteins and genes) into nanoparticles holds promise in fields ranging from catalysis to biomedicine. However, it is challenging to develop a simple and broadly applicable nanoparticle platform that can readily incorporate distinct biomacromolecules without affecting their intrinsic activity. Herein, a metal-phenolic assembly approach is presented whereby diverse functional nanoparticles can be readily assembled in water by combining various synthetic and natural building blocks, including poly(ethylene glycol), phenolic ligands, metal ions, and bioactive macromolecules. The assembly process is primarily mediated by metal-phenolic complexes through coordination and hydrophobic interactions, which yields uniform and spherical nanoparticles (mostly <200 nm), while preserving the function of the incorporated biomacromolecules (siRNA and five different proteins used). The functionality of the assembled nanoparticles is demonstrated through cancer cell apoptosis, RNA degradation, catalysis, and gene downregulation studies. Furthermore, the resulting nanoparticles can be used as building blocks for the secondary engineering of superstructures via templating and cross-linking with metal ions. The bioactivity and versatility of the platform can potentially be used for the streamlined and rational design of future bioactive materials.
Collapse
Affiliation(s)
- Jingqu Chen
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Shuaijun Pan
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Jiajing Zhou
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Zhixing Lin
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yijiao Qu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Agata Glab
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yiyuan Han
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Joseph J Richardson
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
42
|
O'Donnell A, Salimi S, Hart L, Babra T, Greenland B, Hayes W. Applications of supramolecular polymer networks. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2022.105209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
43
|
Liu X, Zhao Z, Wu F, Chen Y, Yin L. Tailoring Hyperbranched Poly(β-amino ester) as a Robust and Universal Platform for Cytosolic Protein Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108116. [PMID: 34894367 DOI: 10.1002/adma.202108116] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/26/2021] [Indexed: 05/24/2023]
Abstract
Cytosolic protein delivery is a prerequisite for protein-based biotechnologies and therapeutics on intracellular targets. Polymers that can complex with proteins to form nano-assemblies represent one of the most important categories of materials, because of the ease of nano-fabrication, high protein loading efficiency, no need for purification, and maintenance of protein bioactivity. Stable protein encapsulation and efficient intracellular liberation are two critical yet opposite processes toward cytosolic delivery, and polymers that can resolve these two conflicting challenges are still lacking. Herein, hyperbranched poly(β-amino ester) (HPAE) with backbone-embedded phenylboronic acid (PBA) is developed to synchronize these two processes, wherein PBA enhanced protein encapsulation via nitrogen-boronate (N-B) coordination while triggered polymer degradation and protein release upon oxidation by H2 O2 in cancer cells. Upon optimization of the branching degree, charge density, and PBA distribution, the best-performing A2-B3-C2-S2 -P2 is identified, which mediates robust delivery of various native proteins/peptides with distinct molecular weights (1.6-430 kDa) and isoelectric points (4.1-10.3) into cancer cells, including enzymes, toxins, antibodies, and CRISPR-Cas9 ribonucleoproteins (RNPs). Moreover, A2-B3-C2-S2 -P2 mediates effective cytosolic delivery of saporin both in vitro and in vivo to provoke remarkable anti-tumor efficacy. Such a potent and universal platform holds transformative potentials for protein pharmaceuticals.
Collapse
Affiliation(s)
- Xun Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Fan Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yongbing Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
44
|
Study for Evaluation of Hydrogels after the Incorporation of Liposomes Embedded with Caffeic Acid. Pharmaceuticals (Basel) 2022; 15:ph15020175. [PMID: 35215288 PMCID: PMC8875116 DOI: 10.3390/ph15020175] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/17/2022] Open
Abstract
Caffeic acid (CA), a phenolic acid, is a powerful antioxidant with proven effectiveness. CA instability gives it limited use, so encapsulation in polymeric nanomaterials has been used to solve the problem but also to obtain topical hydrogel formulas. Two different formulas of caffeic acid liposomes were incorporated into three different formulas of carbopol-based hydrogels. A Franz diffusion cell system was used to evaluate the release of CA from hydrogels. For the viscoelastic measurements of the hydrogels, the equilibrium flow test was used. The dynamic tests were examined at rest by three oscillating tests: the amplitude test, the frequency test and the flow and recovery test. These carbopol gels have a high elasticity at flow stress even at very low polymer concentrations. In the analysis of the texture, the increase of the polymer concentration from 0.5% to 1% determined a linear increase of the values of the textural parameters for hydrogels. The textural properties of 1% carbopol-based hydrogels were slightly affected by the addition of liposomal vesicle dispersion and the firmness and shear work increased with increasing carbomer concentration.
Collapse
|
45
|
Yu H, Palazzolo JS, Zhou J, Hu Y, Niego B, Pan S, Ju Y, Wang TY, Lin Z, Hagemeyer CE, Caruso F. Bioresponsive Polyphenol-Based Nanoparticles as Thrombolytic Drug Carriers. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3740-3751. [PMID: 35019268 DOI: 10.1021/acsami.1c19820] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Thrombolytic (clot-busting) therapies with plasminogen activators (PAs) are first-line treatments against acute thrombosis and ischemic stroke. However, limitations such as narrow therapeutic windows, low success rates, and bleeding complications hinder their clinical use. Drug-loaded polyphenol-based nanoparticles (NPs) could address these shortfalls by delivering a more targeted and safer thrombolysis, coupled with advantages such as improved biocompatibility and higher stability in vivo. Herein, a template-mediated polyphenol-based supramolecular assembly strategy is used to prepare nanocarriers of thrombolytic drugs. A thrombin-dependent drug release mechanism is integrated using tannic acid (TA) to cross-link urokinase-type PA (uPA) and a thrombin-cleavable peptide on a sacrificial mesoporous silica template via noncovalent interactions. Following drug loading and template removal, the resulting NPs retain active uPA and demonstrate enhanced plasminogen activation in the presence of thrombin (1.14-fold; p < 0.05). Additionally, they display lower association with macrophage (RAW 264.7) and monocytic (THP-1) cell lines (43 and 7% reduction, respectively), reduced hepatic accumulation, and delayed blood clearance in vivo (90% clearance at 60 min vs 5 min) compared with the template-containing NPs. Our thrombin-responsive, polyphenol-based NPs represent a promising platform for advanced drug delivery applications, with potential to improve thrombolytic therapies.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jason S Palazzolo
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Jiajing Zhou
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yingjie Hu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Be'eri Niego
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Shuaijun Pan
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ting-Yi Wang
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Zhixing Lin
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christoph E Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
46
|
Sarkar AK, Debnath K, Arora H, Seth P, Jana NR, Jana NR. Direct Cellular Delivery of Exogenous Genetic Material and Protein via Colloidal Nano-Assemblies with Biopolymer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3199-3206. [PMID: 34985241 DOI: 10.1021/acsami.1c22009] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Direct cytosolic delivery of large biomolecules that bypass the endocytic pathways is a promising strategy for therapeutic applications. Recent works have shown that small-molecule, nanoparticle, and polymer-based carriers can be designed for direct cytosolic delivery. It has been shown that the specific surface chemistry of the carrier, nanoscale assembly between the carrier and cargo molecule, good colloidal stability, and low surface charge of the nano-assembly are critical for non-endocytic uptake processes. Here we report a guanidinium-terminated polyaspartic acid micelle for direct cytosolic delivery of protein and DNA. The polymer delivers the protein/DNA directly to the cytosol by forming a nano-assembly, and it is observed that <200 nm size of colloidal assembly with near-zero surface charge is critical for efficient cytosolic delivery. This work shows the importance of size and colloidal property of the nano-assembly for carrier-based cytosolic delivery of large biomolecules.
Collapse
Affiliation(s)
- Ankan Kumar Sarkar
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Koushik Debnath
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, College of Medicine, Chicago, Illinois 60612, United States
| | - Himali Arora
- Cellular and Molecular Neuroscience, National Brain Research Centre, Gurugram, Haryana 122052, India
| | - Pankaj Seth
- Cellular and Molecular Neuroscience, National Brain Research Centre, Gurugram, Haryana 122052, India
| | - Nihar R Jana
- School of Bioscience, Indian Institute of Technology, Kharagpur 721302, India
| | - Nikhil R Jana
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
47
|
Davis HC, Posey ND, Tew GN. Protein Binding and Release by Polymeric Cell-Penetrating Peptide Mimics. Biomacromolecules 2021; 23:57-66. [PMID: 34879198 DOI: 10.1021/acs.biomac.1c00929] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is significant potential in exploiting antibody specificity to develop new therapeutic treatments. However, intracellular protein delivery is a paramount challenge because of the difficulty in transporting large, polar molecules across cell membranes. Cell-penetrating peptide mimics (CPPMs) are synthetic polymers that are versatile materials for intracellular delivery of biological molecules, including nucleic acids and proteins, with superior performance compared to their natural counterparts and commercially available peptide-based reagents. Studies have demonstrated that noncovalent complexation with these synthetic carriers is necessary for the delivery of proteins, but the fundamental interactions dominating CPPM-protein complexation are not well understood. Beyond these interactions, the mechanism of release for many noncovalent carriers is not well established. Herein, interactions expected to be critical in CPPM-protein binding and unbinding were explored, including hydrogen bonding, electrostatics, and hydrophobic interactions. Despite the guanidinium-rich functionality of these polymeric carriers, hydrogen bonding was shown not to be a dominant interaction in CPPM-protein binding. Fluorescence quenching assays were used to decouple the effect of electrostatic and hydrophobic interactions between amphiphilic CPPMs and proteins. Furthermore, by conducting competition assays with other proteins, unbinding of protein cargoes from CPPM-protein complexes was demonstrated and provided insight into mechanisms of protein release. This work offers understanding toward the role of carrier and cargo binding and unbinding in intracellular outcomes. In turn, an improved fundamental understanding of noncovalent polymer-protein complexation will enable more effective methods for intracellular protein delivery.
Collapse
Affiliation(s)
- Hazel C Davis
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Nicholas D Posey
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Gregory N Tew
- Department of Polymer Science & Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States.,Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
48
|
Coiled coil exposure and histidine tags drive function of an intracellular protein drug carrier. J Control Release 2021; 339:248-258. [PMID: 34563592 DOI: 10.1016/j.jconrel.2021.09.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 01/05/2023]
Abstract
In recent years, protein engineering efforts have yielded a diverse set of binding proteins that hold promise for various therapeutic applications. Despite this, their inability to reach intracellular targets limits their applications to cell surface or soluble targets. To address this challenge, we previously reported a protein carrier that binds antibodies and delivers them to therapeutic targets inside cancer cells. This carrier, known as the Hex carrier, is comprised of a self-assembling coiled coil hexamer at the core, with each alpha helix fused to a linker, an antibody binding domain, and a six Histidine-tag (His-tag). In this work, we designed different versions of the carrier to determine the role of each building block in cytosolic protein delivery. We found that increasing exposure of the Hex coiled coil on the carriers, through molecular design or removing antibodies, increased internalization, pointing to a role of the coiled coil in promoting endocytosis. We observed a clear increase in endosomal disruption events when His-tags were present on the carrier relative to when they were removed, due to an endosomal buffering effect. Finally, we found that the antibody binding domains of the Hex carrier could be replaced with monomeric ultra-stable GFP for intracellular delivery and endosomal escape. Our results demonstrate that the Hex coiled coil, in conjunction with His-tags, could be a generalizable vehicle for delivering small and large proteins to intracellular targets. This work also highlights new biological applications for oligomeric coiled coils and shows the direct and quantifiable impact of histidine residues on endosomal disruption. These findings could inform the design of future drug delivery vehicles in applications beyond intracellular protein delivery.
Collapse
|
49
|
Brito J, Hlushko H, Abbott A, Aliakseyeu A, Hlushko R, Sukhishvili SA. Integrating Antioxidant Functionality into Polymer Materials: Fundamentals, Strategies, and Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:41372-41395. [PMID: 34448558 DOI: 10.1021/acsami.1c08061] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
While antioxidants are widely known as natural components of healthy food and drinks or as additives to commercial polymer materials to prevent their degradation, recent years have seen increasing interest in enhancing the antioxidant functionality of newly developed polymer materials and coatings. This paper provides a critical overview and comparative analysis of multiple ways of integrating antioxidants within diverse polymer materials, including bulk films, electrospun fibers, and self-assembled coatings. Polyphenolic antioxidant moieties with varied molecular architecture are in the focus of this Review, because of their abundance, nontoxic nature, and potent antioxidant activity. Polymer materials with integrated polyphenolic functionality offer opportunities and challenges that span from the fundamentals to their applications. In addition to the traditional blending of antioxidants with polymer materials, developments in surface grafting and assembly via noncovalent interaction for controlling localization versus migration of antioxidant molecules are discussed. The versatile chemistry of polyphenolic antioxidants offers numerous possibilities for programmed inclusion of these molecules in polymer materials using not only van der Waals interactions or covalent tethering to polymers, but also via their hydrogen-bonding assembly with neutral molecules. An understanding and rational use of interactions of polyphenol moieties with surrounding molecules can enable precise control of concentration and retention versus delivery rate of antioxidants in polymer materials that are critical in food packaging, biomedical, and environmental applications.
Collapse
Affiliation(s)
- Jordan Brito
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Hanna Hlushko
- Notre Dame Radiation Laboratory, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Ashleigh Abbott
- Department of Materials Science & Engineering, Missouri University of Science & Technology, Rolla, Missouri 65409, United States
| | - Aliaksei Aliakseyeu
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Raman Hlushko
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Svetlana A Sukhishvili
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
50
|
Zhou J, Penna M, Lin Z, Han Y, Lafleur RPM, Qu Y, Richardson JJ, Yarovsky I, Jokerst JV, Caruso F. Robust and Versatile Coatings Engineered via Simultaneous Covalent and Noncovalent Interactions. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Jiajing Zhou
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
- Department of NanoEngineering University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
| | - Matthew Penna
- School of Engineering RMIT University Melbourne Victoria 3001 Australia
| | - Zhixing Lin
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Yiyuan Han
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - René P. M. Lafleur
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Yijiao Qu
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Joseph J. Richardson
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Irene Yarovsky
- School of Engineering RMIT University Melbourne Victoria 3001 Australia
| | - Jesse V. Jokerst
- Department of NanoEngineering University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
- Materials Science and Engineering Program University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
- Department of Radiology University of California San Diego 9500 Gilman Dr. La Jolla CA 92093 USA
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|