1
|
Sudaraka Tennakoon MSBWTMN, Lee KH, Shin HJ. Expression of recombinant swine ferritin heavy chain with enhanced solubility in Escherichia coli and simplified purification of ferritin nanoparticles. Protein Expr Purif 2025; 231:106700. [PMID: 40086537 DOI: 10.1016/j.pep.2025.106700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Ferritin is a versatile biomolecule used in various medical applications such as drug delivery, vaccines, biological imaging, and diagnostics. The purity and concentration of the ferritin nanoparticles are crucial for achieving excellent outcomes. In this study, we expressed and purified the recombinant swine ferritin heavy chain (rsFTH) as a new candidate for recombinant ferritin nanoparticles. We generated two types of plasmids that can express rsFTH in mammalian and prokaryotic systems. The myc-tagged rsFTH expressed in the mammalian system was purified and ferritin nanoparticles were validated using dynamic light scattering (DLS) and transmission electron microscopy (TEM). A prokaryotic expression system was used to produce rsFTH on a large scale. Protein expression was optimized in Escherichia coli BL21 under varying temperatures and IPTG conditions, and solubility was enhanced by incubation at 25 °C for 18-22 h in auto-induction media, resulting in approximately >50 % protein content in the soluble fraction compared with the pellet. Protein purification was achieved using His-tag affinity chromatography and dialysis with Tris-HCl buffer, yielding adequately pure rsFTH without any apparent protein aggregates. SDS-PAGE and Western blot analysis confirmed the expected molecular weight of rsFTH, and Native-PAGE demonstrated polymerization into higher molecular weight forms. Particle size analysis of purified rsFTH revealed a mean diameter of 15.5 nm, with transmission electron microscopy (TEM) imaging confirming spherical ferritin particles with an iron core. These results suggest that rsFTH can be efficiently expressed and purified in both mammalian and bacterial systems, and has potential applications in nanotechnology and biotechnology.
Collapse
Affiliation(s)
| | - Kyoung-Ho Lee
- Laboratory of Infectious diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, South Korea; CellEnVax Co., Ltd, South Korea
| | - Hyun-Jin Shin
- Laboratory of Infectious diseases, College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, South Korea; CellEnVax Co., Ltd, South Korea.
| |
Collapse
|
2
|
Han Z, Guo AX, Luo T, Cai T, Mirkin CA. Biomineralization of semiconductor quantum dots using DNA-functionalized protein nanoreactors. SCIENCE ADVANCES 2025; 11:eadv6906. [PMID: 40333972 PMCID: PMC12057681 DOI: 10.1126/sciadv.adv6906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
Proteins can template the heterogeneous nucleation and growth of size-confined nanocrystals. However, protein-templated mineralization often leads to particles that exhibit low colloidal stability, poor crystal quality, and/or diminished photoluminescence. Here, we report protein cage-spherical nucleic acids (SNAs) that can be used as nanoreactors for quantum dot (QD) synthesis and subsequent intracellular delivery. The resulting QD-SNA structures are monodisperse, colloidally stable, and photoluminescent in aqueous solution. The nanoreactors were prepared using two different proteins (~10 and 12 nanometers in diameter), and CdS, CdSe, and PbSe nanocrystals were synthesized. Moreover, the extent of surface defects and crystallinity depends on the relative concentrations of ionic precursors, which control the growth rate and the number of ionic vacancies. By optimizing conditions, CdS-SNAs that exhibit near-zero reabsorption loss were synthesized. Last, QD-SNAs exhibit enhanced cellular uptake and minimal cytotoxicity when compared to commercial QD-protein conjugates, making them potentially useful in bioimaging and diagnostic applications.
Collapse
Affiliation(s)
- Zhenyu Han
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| | - Allen X. Guo
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Taokun Luo
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| | - Tong Cai
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
3
|
Sanz-Velasco A, Patrian M, Nieddu M, Shen B, Fuenzalida Werner JP, Kostiainen MA, Costa RD, Anaya-Plaza E. Fusing fluorescent proteins and ferritin for protein cage based lighting devices. NANOSCALE 2025; 17:10793-10800. [PMID: 40184033 PMCID: PMC11970472 DOI: 10.1039/d4nr05261g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Ferritin cages are an effective platform to encapsulate and stabilize a range of active cargoes and present a promising stepping stone towards a wide range of applications. They have been explored for optoelectronic applications in combination with fluorescent proteins towards bio-hybrid light-emitting diodes (Bio-HLEDs) only recently. However, protein integration within the cage or coassembled ferritin cages relies on electrostatic interactions and requires the supercharging of the fluorescent protein that easily compromises functionality and stability. To address this limitation, we have developed a fusion protein combining the Thermotoga maritima apoferritin (TmaFt) with a green fluorescent protein named mGreenlantern (mGL). This approach avoids jeopardizing both the cage assembly capability of TmaFt and the photophysical features of mGL. After optimizing the fusion protein mGL-TmaFt with respect to the linker length, assembling efficiency, and mGL payload into the cage (mGL@TmaFt), our findings reveal that they exhibited enhanced thermal and structural stabilities in both solution and when embedded into a polymer matrix. This enables effective mGL shielding, reducing H-transfer deactivation of the chromophore and water-assisted heat transfer across the polymer network. Indeed, the photo-induced heat generation in Bio-HLEDs operating at high currents was significantly reduced, resulting in a 30- and 15-fold higher device stability compared to references with either mGL or mGL-TmaFt proteins, respectively. Overall, this work sets in the potential of protein cage design for photon manipulation in protein lighting devices.
Collapse
Affiliation(s)
- Alba Sanz-Velasco
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| | - Marta Patrian
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Mattia Nieddu
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Boxuan Shen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Juan Pablo Fuenzalida Werner
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Mauri A Kostiainen
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| | - Rubén D Costa
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Eduardo Anaya-Plaza
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| |
Collapse
|
4
|
Ohno N, Karube F, Fujiyama F. Volume electron microscopy for genetically and molecularly defined neural circuits. Neurosci Res 2025; 214:48-55. [PMID: 38914208 DOI: 10.1016/j.neures.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 06/26/2024]
Abstract
The brain networks responsible for adaptive behavioral changes are based on the physical connections between neurons. Light and electron microscopy have long been used to study neural projections and the physical connections between neurons. Volume electron microscopy has recently expanded its scale of analysis due to methodological advances, resulting in complete wiring maps of neurites in a large volume of brain tissues and even entire nervous systems in a growing number of species. However, structural approaches frequently suffer from inherent limitations in which elements in images are identified solely by morphological criteria. Recently, an increasing number of tools and technologies have been developed to characterize cells and cellular components in the context of molecules and gene expression. These advancements include newly developed probes for visualization in electron microscopic images as well as correlative integration methods for the same elements across multiple microscopic modalities. Such approaches advance our understanding of interactions between specific neurons and circuits and may help to elucidate novel aspects of the basal ganglia network involving dopamine neurons. These advancements are expected to reveal mechanisms for processing adaptive changes in specific neural circuits that modulate brain functions.
Collapse
Affiliation(s)
- Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, Japan; Division of Ultrastructural Research, National Institute for Physiological Sciences, Japan.
| | - Fuyuki Karube
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| | - Fumino Fujiyama
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan
| |
Collapse
|
5
|
Cosottini L, Giachetti A, Guerri A, Martinez-Castillo A, Geri A, Zineddu S, Abrescia NGA, Messori L, Turano P, Rosato A. Structural Insight Into a Human H Ferritin@Gold-Monocarbene Adduct: Aurophilicity Revealed in a Biological Context. Angew Chem Int Ed Engl 2025:e202503778. [PMID: 40249912 DOI: 10.1002/anie.202503778] [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: 02/14/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 04/20/2025]
Abstract
Human H ferritin (HuHf) has excellent potential as a nanocarrier for the selective delivery of anticancer metal-based drugs to tumor cells. Here, we addressed the interaction of the gold monocarbene compound Au(NHC)Cl with HuHf by electrospray ionization-mass spectrometry (ESI-MS) measurements, which provide the metalation state of the protein subunits and demonstrate the involvement of protein cysteines in gold binding. The adduct between Au(NHC)Cl and HuHf was studied by cryo-EM measurements, resulting in a high-resolution 3D density map at 1.51 Å. The cryo-EM structure shows a novel tetranuclear gold(I) cluster, located in a surface pocket of each subunit where it is bound to Cys90 and Cys102. The short inter-metal distances are diagnostic of the occurrence of aurophilic interactions. The present work demonstrates the usefulness of cryo-EM to investigate the interactions between metal-based drugs and their protein targets/carriers, also leveraging the strong signal of transition metal ions.
Collapse
Affiliation(s)
- Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
| | - Andrea Giachetti
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), Sesto Fiorentino, FI, 50019, Italy
| | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
| | - Ane Martinez-Castillo
- Structure and Cell Biology of Viruses Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE) Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Andrea Geri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
| | - Stefano Zineddu
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
| | - Nicola G A Abrescia
- Structure and Cell Biology of Viruses Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE) Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, FI, 50019, Italy
| | - Antonio Rosato
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, Sesto Fiorentino, 50019, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, FI, 50019, Italy
| |
Collapse
|
6
|
Szyszka TN, Siddiquee R, Loustau A, Adamson LSR, Rennie C, Huang T, Young R, Care A, Lau YH. High-Fidelity In Vitro Packaging of Diverse Synthetic Cargo into Encapsulin Protein Cages. Angew Chem Int Ed Engl 2025:e202422459. [PMID: 40139971 DOI: 10.1002/anie.202422459] [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: 11/18/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 03/29/2025]
Abstract
Cargo-filled protein cages are powerful tools in biotechnology with demonstrated potential as catalytic nanoreactors and vehicles for targeted drug delivery. While endogenous biomolecules can be packaged into protein cages during their expression and self-assembly inside cells, synthetic cargo molecules are typically incompatible with live cells and must be packaged in vitro. Here, we report a fusion-based in vitro assembly method for packaging diverse synthetic cargo into encapsulin protein cages that outperforms standard in cellulo assembly, producing cages with superior uniformity and thermal stability. Fluorescent dyes, proteins and cytotoxic drug molecules can all be selectively packaged with high efficiency via a peptide-mediated targeting process. The exceptional fidelity and broad compatibility of our in vitro assembly platform enables generalisable access to cargo-filled protein cages that host novel synthetic functionality for diverse biotechnological applications.
Collapse
Affiliation(s)
- Taylor N Szyszka
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
- ARC Centre of Excellence in Synthetic Biology, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Rezwan Siddiquee
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
- ARC Centre of Excellence in Synthetic Biology, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Alex Loustau
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lachlan S R Adamson
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
- ARC Centre of Excellence in Synthetic Biology, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Australian Institute for Microbiology and Infection, Sydney, NSW, 2007, Australia
| | - Tiancheng Huang
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Reginald Young
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Andrew Care
- ARC Centre of Excellence in Synthetic Biology, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yu Heng Lau
- School of Chemistry, The University of Sydney, Camperdown, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
- ARC Centre of Excellence in Synthetic Biology, The University of Sydney, Camperdown, NSW, 2006, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, NSW, 2006, Australia
| |
Collapse
|
7
|
Xia B, Feng H, Jiang X, Guo J, Lin K, Zhang W, Xing F, Cao L, Li Y, Zhang H, Zhang X, Li W, Yu F. Development of chimeric Nanobody-Granzyme B functionalized ferritin nanoparticles for precise tumor therapy. Pharmacol Res 2025; 213:107628. [PMID: 39880067 DOI: 10.1016/j.phrs.2025.107628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/25/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
T-cell lymphomas (TCLs) are heterogeneous malignancies with limited treatment options and poor outcomes. The efficacy of traditional T-cell therapies, including chimeric antigen receptor (CAR) T cells, is often constrained by immunosuppressive factors and the tumor microenvironment. On the other hand, although direct Granzyme B (GrB) administration can effectively induce tumor cell apoptosis, it lacks universal tumor targeting and efficient cellular entry mechanisms. To address these limitations, we developed a novel nanoparticle-based therapy for the precise targeting of TCL tumor cells and the delivery of GrB. We fused nanobody (Nb) targeting CD30 and CD5 with GrB and coupled them to human ferritin (h-HFn) using the Gv/Sd system, creating a novel therapeutic nanoparticle named BiCD30/5-GF, which specifically targets CD30 and CD5 receptors on TCL tumor cells. The Nb-GrB conjugation enhances tumor targeting, while a Gv/Sd linker coupled to h-HFn further improves cellular transport and targeting. Additionally, the multimerization of GrB enhances its effectiveness. These nanoparticles demonstrated superior binding affinity and cytotoxicity in vitro compared to conventional treatments. In vivo studies on tumor-bearing mice showed significant tumor suppression and prolonged survival following treatment with BiCD30/5-GF nanoparticles. We also extended similar nanoparticle strategies for gastric cancer therapy, targeting FGFR4-expressing tumor cells. Our findings highlight the potential of engineered nanoparticles as effective and targeted therapeutic agents across various tumor types, offering promising prospects for clinical translation in cancer treatment.
Collapse
Affiliation(s)
- Baijin Xia
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China; Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Huolun Feng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xinmiao Jiang
- Lymphoma Department, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jialing Guo
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Keming Lin
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wenxing Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fan Xing
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Yong Li
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Xu Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Wenyu Li
- Lymphoma Department, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
8
|
Xia Y, Li X, Huang F, Wu Y, Liu J, Liu J. Design and advances in antioxidant hydrogels for ROS-induced oxidative disease. Acta Biomater 2025; 194:80-97. [PMID: 39900274 DOI: 10.1016/j.actbio.2025.01.057] [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: 09/30/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Reactive oxygen species (ROS) play a crucial role in human physiological processes, but oxidative stress caused by excessive ROS may lead to a variety of acute and chronic diseases. Despite the development of various strategies and biomaterials, an efficiently and broadly applied method for treatment of ROS-induced oxidative disease remains a bottleneck. Aiming to improve the local oxidative stress environment, numerous bioactive hydrogels with antioxidant properties have emerged and are proven to quickly and continuously eliminate excessive ROS. To deeply understand the design principles and applications of antioxidant hydrogels is highly beneficial for designing antioxidant hydrogels for treatment of oxidative disease. This review provides a detailed summary of recent advances in design and applications of antioxidant hydrogels for various ROS-induced oxidative diseases. In this review, the kinds of antioxidant components in antioxidant hydrogels are outlined in detail. Additionally, the crosslinking methods and the biomedical applications of antioxidant hydrogels are widely summarized and discussed, especially focusing on their usage in different types of diseases and the attention given to the treatment of diseases such as skin wounds, myocardial infarction, and osteoarthritis. Finally, the future development direction of antioxidant hydrogel is further proposed. STATEMENT OF SIGNIFICANCE: Oxidative stress is a pivotal biochemical process that plays a critical role in cellular homeostasis. Excessive cellular oxidative stress triggers an inflammatory response, which is implicated in a spectrum of associated diseases. Given the critical need for managing oxidative stress, antioxidant therapies have become a vital focus in medical research. Hydrogels have garnered substantial interest among biomaterial scientists due to their hydrophilic nature and biocompatibility. The review delves into the realm of antioxidant hydrogels, encompassing the classification of antioxidant components, the synthesis and fabrication of hydrogels, and a comprehensive overview of the biological applications and challenges of these antioxidant hydrogels. Aiming to provide new perspectives for researchers in developing cutting-edge therapeutic approaches that leverage antioxidant hydrogels.
Collapse
Affiliation(s)
- Yi Xia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanhao Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
9
|
Zhang B, Fan K. Design and application of ferritin-based nanomedicine for targeted cancer therapy. Nanomedicine (Lond) 2025; 20:481-500. [PMID: 39895329 PMCID: PMC11875477 DOI: 10.1080/17435889.2025.2459056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
Owing to its unique structure and favorable biocompatibility, ferritin has been widely studied as a promising drug carrier over the past two decades. Since the identification of its inherent tumor-targeting property due to unique recognition ablity of the transferrin receptor 1 (TfR1), ferritin-based nanomedicine has attracted widespread attention and triggered a research surge in the field of targeted cancer therapy. Along with progress in structure studies and modification technology, diverse strategies have been carried out to equip ferritin with on-demand functions, further improving the antitumor efficacy and in vivo safety of ferritin-based cancer therapy. In this review, we highlight the structure-based rational design of ferritin and summarize the design strategies in detail from two main perspectives: multifunctional modification and drug loading. In particular, the critical issues that need attention in the design are discussed in depth. Furthermore, we provide an overview of the latest advances in the application of ferritin-based nanomedicines in chemotherapy, phototherapy and immunotherapy, with particular emphasis on emerging therapeutic approaches among these therapies.
Collapse
Affiliation(s)
- Baoli Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Yoshida E. Protein Cage-like Vesicles Fabricated via Polymerization-Induced Microphase Separation of Amphiphilic Diblock Copolymers. MATERIALS (BASEL, SWITZERLAND) 2025; 18:727. [PMID: 39942392 PMCID: PMC11820364 DOI: 10.3390/ma18030727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025]
Abstract
Highly symmetric protein cages represent one of the most artistic architectures formed by biomolecules. However, the underlying reasons for the formation of some of these architectures remain unknown. The present study aims to investigate the significance behind their morphological formation by fabricating protein cage-like vesicles using a synthetic polymer. The vesicles were synthesized by combining polymerization-induced self-assembly (PISA) with polymerization-induced microphase separation (PIMS), employing an amphiphilic poly(methacrylic acid)-block-poly(n-butyl methacrylate-random-cyclohexyl methacrylate-random-methacrylic acid) diblock copolymer, PMAA-b-P(BMA-r-CMA-r-MAA). The copolymer, with a 60 mol% molar ratio of CMA to the BMA units, produced clathrin-like vesicles with angular windows in their shell, resulting from the segregation of the hard CMA units from the soft BMA matrix in the hydrophobic phase of the vesicle. These vesicles were highly stable against rising temperatures. In contrast, the vesicles with a 30 mol% CMA ratio dissociated upon heating to 50 °C into triskelion-like segments due to intramolecular microphase separation. These findings indicate that designing synthetic polymers can mimic living organ morphologies, aiding in elucidating their morphological significance and inspiring the development of new materials utilizing these morphologies.
Collapse
Affiliation(s)
- Eri Yoshida
- Department of Applied Chemistry and Life Science, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi 441-8580, Japan
| |
Collapse
|
11
|
Amrita, Chakraborti S, Dey S. Physicochemical features of subunit interfaces and their role in self-assembly across the ferritin superfamily. Structure 2025; 33:401-415.e2. [PMID: 39740669 DOI: 10.1016/j.str.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/29/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Ferritins are ubiquitous and play a critical role in iron homeostasis. They are classified into four main subfamilies: classical, bacterial, bacterioferritin, and Dps. These are characterized by subunits with a four-helical bundle domain and interact through three distinct regions-one antiparallel interface (IntA) and two perpendicular interfaces (IntB and IntC), collectively forming a cage-like structure. Here, we attempt to characterize the variability of these interfaces across subfamilies. We found that IntA is essential for the dimeric unit assembly and is likely to assemble first, followed by the smaller interfaces of IntB and IntC (in any order), which are crucial for cage formation. These interfaces are unique in that they are less packed, although chemically stable, and their size lies between that of protein-protein complex and obligate homodimers. This study provides a detailed exploration of the ferritin interfaces, offering insights into their assembly and their importance as carrier proteins.
Collapse
Affiliation(s)
- Amrita
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Jodhpur, Rajasthan, India
| | - Soumyananda Chakraborti
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Dist.-Medchal, 500 078, Hyderabad, Telangana, India.
| | - Sucharita Dey
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342030, Jodhpur, Rajasthan, India.
| |
Collapse
|
12
|
Kazakov EP, Kireev II, Golyshev SA. Techniques for Selective Labeling of Molecules and Subcellular Structures for Cryo-Electron Tomography. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:173-187. [PMID: 40254397 DOI: 10.1134/s0006297924604015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 04/22/2025]
Abstract
Electron microscopy (EM) is one of the most efficient methods for studying the fine structure of cells with a resolution thousands of times higher than that of visible light microscopy. The most advanced implementation of electron microscopy in biology is EM tomography of samples stabilized by freezing without water crystallization (cryoET). By circumventing the drawbacks of chemical fixation and dehydration, this technique allows investigating cellular structures in three dimensions at the molecular level, down to resolving individual proteins and their subdomains. However, the problem of efficient identification and localization of objects of interest has not yet been solved, thus limiting the range of targets to easily recognizable or abundant subcellular components. Labeling techniques provide the only way for locating the subject of investigation in microscopic images. CryoET imposes conflicting demands on the labeling system, including the need to introduce into a living cell the particles composed of substances foreign to the cellular chemistry that have to bind to the molecule of interest without disrupting its vital functions and physiology of the cell. This review examines both established and prospective methods for selective labeling of proteins and subcellular structures aimed to enable their localization in cryoET images.
Collapse
Affiliation(s)
- Evgeny P Kazakov
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Igor I Kireev
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
- Department of Cell Biology and Histology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Sergei A Golyshev
- Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
13
|
Sen S, Thaker A, Haymaker A, Williams D, Chiu PL, Nannenga BL. Observation of the Protein-Inorganic Interface of Ferritin by Cryo-Electron Microscopy. J Am Chem Soc 2025; 147:3333-3340. [PMID: 39815632 DOI: 10.1021/jacs.4c13873] [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: 01/18/2025]
Abstract
Visualizing the structure of the protein-inorganic interface is critically important for a more complete understanding of biomineralization. Unfortunately, there are limited approaches for the direct and detailed study of biomolecules that interact with inorganic materials. Here, we use single-particle cryo-electron microscopy (cryo-EM) to study the protein-nanoparticle (NP) interactions of human light chain ferritin and visualize the high-resolution details of the protein-inorganic interface. In this work, we determined the 2.85 Å structure of human light chain ferritin bound to its native iron oxide NP substrate. The resulting cryo-EM maps confirmed and enhanced previously proposed interactions of the protein with the material along the B-helix and revealed new interaction at the C-terminus of light chain ferritin. This work sheds new light on the mechanisms of ferritin biomineralization and further demonstrates the application of cryo-EM for the study of protein-inorganic systems.
Collapse
Affiliation(s)
- Sagnik Sen
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, Arizona 85281, United States
| | - Amar Thaker
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, Arizona 85281, United States
| | - Alison Haymaker
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, Arizona 85281, United States
| | - Dewight Williams
- John M. Cowley Center for High Resolution Electron Microscopy, Arizona State University, Tempe, Arizona 85287, United States
| | - Po-Lin Chiu
- Biodesign Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, Arizona 85281, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Brent L Nannenga
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Center for Applied Structural Discovery, Biodesign Institute, Arizona State University, 727 East Tyler Street, Tempe, Arizona 85281, United States
| |
Collapse
|
14
|
Oh HJ, Lee Y, Hwang H, Hong K, Choi H, Kang JY, Jung Y. Size-controlled assembly of phase separated protein condensates with interfacial protein cages. Nat Commun 2025; 16:1009. [PMID: 39856105 PMCID: PMC11760349 DOI: 10.1038/s41467-025-56391-y] [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: 04/10/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Phase separation of specific proteins into liquid-like condensates is a key mechanism for forming membrane-less organelles, which organize diverse cellular processes in space and time. These protein condensates hold immense potential as biomaterials capable of containing specific sets of biomolecules with high densities and dynamic liquid properties. Despite their appeal, methods to manipulate protein condensate materials remain largely unexplored. Here, we present a one-pot assembly method to assemble coalescence-resistant protein condensates, ranging from a few μm to 100 nm in sizes, with surface-stabilizing protein cages. We discover that large protein cages (~30 nm), finely tuned to interact with condensates, efficiently localize on condensate surfaces and prevent the merging (coalescence) of condensates during phase separation. We precisely control condensate diameters by modulating condensate/cage ratios. In addition, the 3D structures of intact protein condensates with interfacial cages are visualized with cryo-electron tomography (ET). This work offers a versatile platform for designing size-controlled, surface-engineered protein condensate materials.
Collapse
Affiliation(s)
- Hyeok Jin Oh
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yongsuk Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Haerang Hwang
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Kibeom Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyeongjoo Choi
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jin Young Kang
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yongwon Jung
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
15
|
Otange B, Katenkamp T, Böhler H, Rütten M, Lang L, Schulz F, Parak WJ, Beck T. Systematic probing of protein adsorption on protein-based nanoparticles in dependence of the particle surface charge. NANOSCALE 2025; 17:1997-2003. [PMID: 39641532 DOI: 10.1039/d4nr04069d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Understanding protein adsorption on the surface of nanoparticles (NPs) is crucial for determining their behavior in biological environments. Early research in this field faced challenges in producing high-quality NPs. Advancements in NP fabrication now allow for precise modifications of specific parameters, such as zeta potential. However, creating a series of NPs where only one parameter, such as surface charge, is independently varied remains challenging due to concurrent alterations in other properties. In this study, we address these challenges using the ferritin nanocage (Ftn) as a model system for NPs. By modifying only a few amino acids on the outer surface of Ftn, we produce NPs with highly defined properties, focusing solely on variations in surface charge. This approach enables us to generate a controlled series of protein-based nanocages, labeled with fluorophores inside the nanocage. We utilize fluorescent correlation spectroscopy (FCS) to investigate the adsorption of bovine serum albumin (BSA) on these NPs, analyzing the dependence of BSA binding on surface charge. This fundamental study enhances our understanding of the driving forces behind protein adsorption, contributing valuable insights into the design of NPs for biomedical applications.
Collapse
Affiliation(s)
- Ben Otange
- Institute for Nanostructure and Solid State Physics, University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Tobias Katenkamp
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany.
| | - Hendrik Böhler
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany.
| | - Michael Rütten
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany.
| | - Laurin Lang
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany.
| | - Florian Schulz
- Institute for Nanostructure and Solid State Physics, University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Wolfgang J Parak
- Institute for Nanostructure and Solid State Physics, University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
- Hamburg Centre for Ultrafast Imaging, University of Hamburg, Luruper Chaussee 149, Hamburg, Germany
| | - Tobias Beck
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany.
- Hamburg Centre for Ultrafast Imaging, University of Hamburg, Luruper Chaussee 149, Hamburg, Germany
| |
Collapse
|
16
|
Zhang R, Shen Y, Zhou X, Li J, Zhao H, Zhang Z, Zhao J, Jin H, Guo S, Ding H, Nie G, Zhang Z, Wang Y, Yan X, Fan K. Hypoxia-tropic delivery of nanozymes targeting transferrin receptor 1 for nasopharyngeal carcinoma radiotherapy sensitization. Nat Commun 2025; 16:890. [PMID: 39837820 PMCID: PMC11751138 DOI: 10.1038/s41467-025-56134-z] [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: 12/20/2023] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC), a malignancy highly prevalent in East and Southeast Asia, is primarily treated with radiotherapy (RT). However, hypoxia-induced radioresistance presents a significant challenge. Nanozymes, nanomaterials with catalase-like activity, have emerged as a promising strategy for radiosensitization by converting elevated hydrogen peroxide in the tumor microenvironment into oxygen. Despite their potential, effectively targeting hypoxic lesions has been difficult. Here, we identify transferrin receptor 1 (TfR1) as an upregulated target in NPC, with its expression levels positively correlated with hypoxia. Human heavy-chain ferritin, a specific ligand of TfR1, selectively recognizes hypoxic NPC lesions in preclinical models. Based on these findings, we design a hypoxia-targeted nanozyme by loading platinum nanoparticles into ferritin. This nanozyme exhibits enhanced catalase-like activity and effectively alleviates tumor hypoxia in NPC xenografts. When combined with RT, a single injection of the nanozyme significantly inhibits tumor growth and prolongs mouse survival, outperforming sodium glycididazole, a clinically used radiosensitizer. In summary, our findings highlight TfR1 as an accessible cell surface target in hypoxic NPC lesions. The nanozyme targeting TfR1 holds promise for enhancing the therapeutic effectiveness of RT in NPC through an in situ oxygen-generation mechanism.
Collapse
Affiliation(s)
- Ruofei Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yanfang Shen
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Jianru Li
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hanqing Zhao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Zixia Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Shuanshuan Guo
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Hui Ding
- Shenzhen Key Laboratory of nanozymes and Translational Cancer Research, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Guohui Nie
- Shenzhen Key Laboratory of nanozymes and Translational Cancer Research, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhe Zhang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Ying Wang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| |
Collapse
|
17
|
Moglia I, Santiago M, Arellano A, Salazar Sandoval S, Olivera-Nappa Á, Kogan MJ, Soler M. Synthesis of dumbbell-like heteronanostructures encapsulated in ferritin protein: Towards multifunctional protein based opto-magnetic nanomaterials for biomedical theranostic. Colloids Surf B Biointerfaces 2024; 245:114332. [PMID: 39486373 DOI: 10.1016/j.colsurfb.2024.114332] [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: 04/10/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
Dumbbell-like hetero nanostructures based on gold and iron oxides is a promising material for biomedical applications, useful as versatile theranostic agents due the synergistic effect of their optical and magnetic properties. However, achieving precise control on their morphology, size dispersion, colloidal stability, biocompatibility and cell targeting remains as a current challenge. In this study, we address this challenge by employing biomimetic routes, using ferritin protein nanocages as template for these nanoparticles' synthesis. We present the development of an opto-magnetic nanostructures using the ferritin protein, wherein gold and iron oxide nanostructures were produced within its cavity. Initially, we investigated the synthesis of gold nanostructures within the protein, generating clusters and plasmonic nanoparticles. Subsequently, we optimized the conditions for the superparamagnetic nanoparticles synthesis through controlled iron oxidation, thereby enhancing the magnetic properties of the resulting system. Finally, we produce magnetic nanoparticles in the protein with gold clusters, achieving the coexistence of both nanostructures within a single protein molecule, a novel material unprecedented to date. We observed that factors such as temperature, metal/protein ratios, pH, dialysis, and purification processes all have an impact on protein recovery, loading efficiency, morphology, and nanoparticle size. Our findings highlight the development of ferritin-based nanomaterials as versatile platforms for potential biomedical use as multifunctional theranostic agents.
Collapse
Affiliation(s)
- Italo Moglia
- Departamento de Química, Facultad de Ciencias Naturales, Matemáticas y del Medioambiente, Universidad Tecnológica Metropolitana-UTEM, Chile.
| | | | - Andreas Arellano
- Instituto Universitario de Investigación y Desarrollo Tecnológico-IDT, Universidad Tecnológica Metropolitana-UTEM, Chile; Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile; Millennium Nucleus in NanoBioPhysics-N2BP, Chile
| | | | - Álvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering-CEBiB, Chile; Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Chile
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile; Advanced Center for Chronic Diseases-ACCDiS, Chile
| | - Mónica Soler
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Chile
| |
Collapse
|
18
|
Cosottini L, Geri A, Ghini V, Mannelli M, Zineddu S, Di Paco G, Giachetti A, Massai L, Severi M, Gamberi T, Rosato A, Turano P, Messori L. Unlocking the Power of Human Ferritin: Enhanced Drug Delivery of Aurothiomalate in A2780 Ovarian Cancer Cells. Angew Chem Int Ed Engl 2024; 63:e202410791. [PMID: 38949226 DOI: 10.1002/anie.202410791] [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: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
Aurothiomalate (AuTM) is an FDA-approved antiarthritic gold drug with unique anticancer properties. To enhance its anticancer activity, we prepared a bioconjugate with human apoferritin (HuHf) by attaching some AuTM moieties to surface protein residues. The reaction of apoferritin with excess AuTM yielded a single adduct, that was characterized by ESI MS and ICP-OES analysis, using three mutant ferritins and trypsinization experiments. The adduct contains ~3 gold atoms per ferritin subunit, arranged in a small cluster bound to Cys90 and Cys102. MD simulations provided a plausible structural model for the cluster. The adduct was evaluated for its pharmacological properties and was found to be significantly more cytotoxic than free AuTM against A2780 cancer cells mainly due to higher gold uptake. NMR-metabolomics showed that AuTM bound to HuHf and free AuTM induced qualitatively similar changes in treated cancer cells, indicating that the effects on cell metabolism are approximately the same, in agreement with independent biochemical experiments. In conclusion, we have demonstrated here that a molecularly precise bioconjugate formed between AuTM and HuHf exhibits anticancer properties far superior to the free drug, while retaining its key mechanistic features. Evidence is provided that human ferritin can serve as an excellent carrier for this metallodrug.
Collapse
Affiliation(s)
- Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Andrea Geri
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Veronica Ghini
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Stefano Zineddu
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Giorgio Di Paco
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Andrea Giachetti
- Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP), 50019, Sesto Fiorentino, FI, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Mirko Severi
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Antonio Rosato
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
- Magnetic Resonance Center, University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
- Magnetic Resonance Center, University of Florence, 50019, Sesto Fiorentino, FI, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, 50019, Sesto Fiorentino, FI, Italy
| |
Collapse
|
19
|
Kwon S, Andreas MP, Giessen TW. Pore Engineering as a General Strategy to Improve Protein-Based Enzyme Nanoreactor Performance. ACS NANO 2024; 18:25740-25753. [PMID: 39226211 PMCID: PMC11971687 DOI: 10.1021/acsnano.4c08186] [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: 09/05/2024]
Abstract
Enzyme nanoreactors are nanoscale compartments consisting of encapsulated enzymes and a selectively permeable barrier. Sequestration and colocalization of enzymes can increase catalytic activity, stability, and longevity, highly desirable features for many biotechnological and biomedical applications of enzyme catalysts. One promising strategy to construct enzyme nanoreactors is to repurpose protein nanocages found in nature. However, protein-based enzyme nanoreactors often exhibit decreased catalytic activity, partially caused by a mismatch of protein shell selectivity and the substrate requirements of encapsulated enzymes. No broadly applicable and modular protein-based nanoreactor platform is currently available. Here, we introduce a pore-engineered universal enzyme nanoreactor platform based on encapsulins-microbial self-assembling protein nanocompartments with programmable and selective enzyme packaging capabilities. We structurally characterize our protein shell designs via cryo-electron microscopy and highlight their polymorphic nature. Through fluorescence polarization assays, we show their improved molecular flux behavior and highlight their expanded substrate range via a number of proof-of-concept enzyme nanoreactor designs. This work lays the foundation for utilizing our encapsulin-based nanoreactor platform for diverse future biotechnological and biomedical applications.
Collapse
Affiliation(s)
- Seokmu Kwon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Michael P Andreas
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Tobias W Giessen
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
20
|
Ricci C, Abbandonato G, Giannangeli M, Matthews L, Almásy L, Sartori B, Podestà A, Caselli A, Boffi A, Thiel G, Del Favero E, Moroni A. Ferritin at different iron loading: From biological to nanotechnological applications. Int J Biol Macromol 2024; 276:133812. [PMID: 39032902 DOI: 10.1016/j.ijbiomac.2024.133812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
The characterization of the structure of ferritin in solution and the arrangement of iron stored in its cavity are intriguing subjects for both cell biology and applied science, since the protein structure, stability, and easiness of production make it an ideal tool for biomedical applications. We characterized the ferritin structure over a wide range of iron loadings by visible light, X-ray, and neutron scattering techniques. We found that the arrangement of iron ions inside the protein cage resulted in a more disposable arrangement at lower loading factors and then in a crystalline structure. At very high iron content the inner core is composed of magnetite more than ferrihydrite, and the shell of the protein is elastically deformed by the iron crystal growth in an ellipsoidal arrangement. The application of an external radiofrequency (RF) magnetic field affected ferritins at low iron loading factors. Notably the RF modified the iron disposition towards a more dispersed arrangement. The structural characterization of the ferritin at different LFs and in presence of magnetic fields provides useful insights into their physiological behaviour and can help in the design and fine-tuning of ferritin-based nanosystems for biotechnological applications.
Collapse
Affiliation(s)
| | | | | | - Lauren Matthews
- ESRF, The European Synchrotron, 71 avenue des Martyrs, 38043 Grenoble, France
| | - László Almásy
- HUN-REN Centre for Energy Research, POB 49, Budapest 1525, Hungary
| | - Barbara Sartori
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9/4, Graz, Austria
| | - Alessandro Podestà
- Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | | | - Alberto Boffi
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - Gerhard Thiel
- Department of Biosciences, University of Milan, Milan, Italy
| | | | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Raut RK, Bhattacharyya G, Behera RK. Gastric stability of bare and chitosan-fabricated ferritin and its bio-mineral: implication for potential dietary iron supplements. Dalton Trans 2024; 53:13815-13830. [PMID: 39109655 DOI: 10.1039/d4dt01839g] [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: 08/21/2024]
Abstract
Iron deficiency anaemia (IDA), the most widespread nutritional disorder, is a persistent global health issue affecting millions, especially in resource-limited geographies. Oral iron supplementation is usually the first choice for exogenous iron administration owing to its convenience, effectiveness and low cost. However, commercially available iron supplementations are often associated with oxidative stress, gastrointestinal side effects, infections and solubility issues. Herein, we aim to address these limitations by employing ferritin proteins-self-assembled nanocaged architectures functioning as a soluble cellular iron repository-as a non-toxic and biocompatible alternative. Our in vitro studies based on PAGE and TEM indicate that bare ferritin proteins are resistant to gastric conditions but their cage integrity is compromised under longer incubation periods and at higher concentrations of pepsin, which is a critical component of gastric juice. To ensure the safe delivery of encapsulated iron cargo, with minimal cage disintegration/degradation and iron leakage along the gastrointestinal tract, we fabricated the surface of ferritin with chitosan. Further, the stoichiometry and absorptivity of iron-chelator complexes at both gastric and circumneutral pH were estimated using Job's plot. Unlike bipyridyl, deferiprone exhibited pH dependency. In vitro kinetics was studied to evaluate iron release from bare and chitosan-fabricated ferritins employing both reductive (in the presence of ascorbate and bipyridyl) and non-reductive (direct chelation by deferiprone) pathways to determine their bio-mineral stabilities. Chitosan-decorated ferritin displayed superior cage integrity and iron retention capability over bare ferritin in simulated gastric fluid. The ability of ferritins to naturally facilitate controlled iron release in conjugation with enteric coating provided by chitosan may mitigate the aforementioned side effects and enhance iron absorption in the intestine. The results of the current study could pave the way for the development of an oral formulation based on ferritin-caged iron bio-mineral that can be a promising alternative for the treatment of IDA, offering better therapeutic outcomes.
Collapse
Affiliation(s)
- Rohit Kumar Raut
- Department of Chemistry, National Institute of Technology, Rourkela - 769008, Odisha, India.
| | - Gargee Bhattacharyya
- Department of Chemistry, National Institute of Technology, Rourkela - 769008, Odisha, India.
| | - Rabindra K Behera
- Department of Chemistry, National Institute of Technology, Rourkela - 769008, Odisha, India.
| |
Collapse
|
22
|
Zhang X, Zhang B, Zhang Y, Ding Y, Zhang Z, Liu Q, Yang Z, Wang L, Gao J. Copper-Induced Supramolecular Peptide Assemblies for Multi-Pathway Cell Death and Tumor Inhibition. Angew Chem Int Ed Engl 2024; 63:e202406602. [PMID: 38837577 DOI: 10.1002/anie.202406602] [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: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Although self-assembly has emerged as an effective tool for fabricating biomaterials, achieving precise control over the morphologies and functionalities of the resultant assemblies remains an ongoing challenge. Inspired by the copper peptide naturally present in human plasma, in this study, we designed a synthetic precursor, FcGH. FcGH can self-assemble via two distinct pathways: spontaneous and Cu2+-induced. These two assembly pathways enabled the formation of assemblies with tunable morphologies by adjusting the amount of added Cu2+. We found that the nanoparticles formed by Cu2+-induced self-assembly exhibited a significantly higher cellular uptake efficiency than the wormlike fibers formed spontaneously. Moreover, this Cu2+-induced assembly process occurred spontaneously at a 1 : 1 molar ratio of Cu2+ to FcGH, avoiding the excessive use of Cu2+ and a tedious preparation procedure. By co-assembling with 10-hydroxycamptothecin (HCPT)-conjugated FcGH, Cu2+-induced supramolecular nanodrugs elicited multiple cell death modalities in cancer cells with elevated immunogenicity, enhancing the therapeutic effect compared to free HCPT. This study highlights Cu2+-induced self-assembly as an efficient tool for directing the assembly of nanodrugs and for synergistic tumor therapy.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Buyue Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Yinghao Ding
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Zhenghao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University Xuzhou, Jiangsu, 221002, China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| |
Collapse
|
23
|
Sun R, Lim S. Ferritin cages as building blocks for higher-order assembly through copper-sulfur bonds for HER analysis. RSC Adv 2024; 14:24791-24796. [PMID: 39114434 PMCID: PMC11305402 DOI: 10.1039/d4ra02931c] [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: 04/20/2024] [Accepted: 05/30/2024] [Indexed: 08/10/2024] Open
Abstract
Higher-order assembly of ferritins has been achieved on copper substrate by introducing cysteines on their surfaces with thiol groups as the active moiety. To elucidate the assembly mechanism, Raman spectroscopy was utilized to characterize the interaction between the copper substrate and the modified ferritin, AfFtnAA/E94C. The resulting higher-order architecture shows enhanced hydrogen evolution reaction activity.
Collapse
Affiliation(s)
- Ruoxuan Sun
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 70 Nanyang Drive 637457 Singapore
| | - Sierin Lim
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 70 Nanyang Drive 637457 Singapore
| |
Collapse
|
24
|
Sheng Y, Chen Z, Cherrier MV, Martin L, Bui TTT, Li W, Lynham S, Nicolet Y, Ebrahimi KH. A Versatile Virus-Mimetic Engineering Approach for Concurrent Protein Nanocage Surface-Functionalization and Cargo Encapsulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310913. [PMID: 38726952 DOI: 10.1002/smll.202310913] [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: 11/26/2023] [Revised: 04/26/2024] [Indexed: 08/02/2024]
Abstract
Naturally occurring protein nanocages like ferritin are self-assembled from multiple subunits. Because of their unique cage-like structure and biocompatibility, there is a growing interest in their biomedical use. A multipurpose and straightforward engineering approach does not exist for using nanocages to make drug-delivery systems by encapsulating hydrophilic or hydrophobic drugs and developing vaccines by surface functionalization with a protein like an antigen. Here, a versatile engineering approach is described by mimicking the HIV-1 Gap polyprotein precursor. Various PREcursors of nanoCages (PREC) are designed and created by linking two ferritin subunits via a flexible linker peptide containing a protease cleavage site. These precursors can have additional proteins at their N-terminus, and their protease cleavage generates ferritin-like nanocages named protease-induced nanocages (PINCs). It is demonstrated that PINC formation allows concurrent surface decoration with a protein and hydrophilic or hydrophobic drug encapsulation up to fourfold more than the amount achieved using other methods. The PINCs/Drug complex is stable and efficiently kills cancer cells. This work provides insight into the precursors' design rules and the mechanism of PINCs formation. The engineering approach and mechanistic insight described here will facilitate nanocages' applications in drug delivery or as a platform for making multifunctional therapeutics like mosaic vaccines.
Collapse
Affiliation(s)
- Yujie Sheng
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Zilong Chen
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Mickael V Cherrier
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Lydie Martin
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Tam T T Bui
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, SE11UL, UK
| | - Wei Li
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Steven Lynham
- Proteomics Core Facility, James Black Centre, King's College London, London, SE5 9NU, UK
| | - Yvain Nicolet
- Univ. Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, Grenoble, CS 10090, France
| | - Kourosh H Ebrahimi
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| |
Collapse
|
25
|
Bhatt S, Dasgupta S, Tupe C, Prashar C, Adhikari U, Pandey KC, Kundu S, Chakraborti S. Antimalarial Delivery with a Ferritin-Based Protein Cage: A Step toward Developing Smart Therapeutics against Malaria. Biochemistry 2024; 63:1738-1751. [PMID: 38975628 DOI: 10.1021/acs.biochem.3c00692] [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: 07/09/2024]
Abstract
Over the past two decades, the utilization of protein cages has witnessed exponential growth driven by their extensive applications in biotechnology and therapeutics. In the context of the recent Covid-19 pandemic, protein-cage-based scaffolds played a pivotal role in vaccine development. Beyond vaccines, these protein cages have proven valuable in diverse drug delivery applications thanks to their distinctive architecture and structural stability. Among the various types of protein cages, ferritin-based cages have taken the lead in drug delivery applications. This is primarily attributed to their ease of production, exceptional thermal stability, and nontoxic nature. While ferritin-based cages are commonly employed in anticancer drug delivery and contrast agent delivery, their efficacy in malarial drug delivery had not been explored until this study. In this investigation, several antimalarial drugs were encapsulated within horse spleen ferritin, and the binding and loading processes were validated through both experimental and computational techniques. The data unequivocally demonstrate the facile incorporation of antimalarial drugs into ferritin without disrupting its three-dimensional structure. Computational docking and molecular dynamics simulations were employed to pinpoint the precise location of the drug binding site within ferritin. Subsequent efficacy testing on Plasmodium revealed that the developed nanoconjugate, comprising the drug-ferritin conjugate, exhibited significant effectiveness in eradicating the parasite. In conclusion, the findings strongly indicate that ferritin-based carrier systems hold tremendous promise for the future of antimalarial drug delivery, offering high selectivity and limited side effects.
Collapse
Affiliation(s)
- Shruti Bhatt
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Subrata Dasgupta
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Chiging Tupe
- ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India
| | - Cherish Prashar
- ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India
| | - Utpal Adhikari
- National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Kailash C Pandey
- ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K K Birla Goa Campus, Goa 403726, India
| | - Soumyananda Chakraborti
- ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
- Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Hyderabad 500078, India
| |
Collapse
|
26
|
Xu M, Wei S, Duan L, Ji Y, Han X, Sun Q, Weng L. The recent advancements in protein nanoparticles for immunotherapy. NANOSCALE 2024; 16:11825-11848. [PMID: 38814163 DOI: 10.1039/d4nr00537f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In recent years, the advancement of nanoparticle-based immunotherapy has introduced an innovative strategy for combatting diseases. Compared with other types of nanoparticles, protein nanoparticles have obtained substantial attention owing to their remarkable biocompatibility, biodegradability, ease of modification, and finely designed spatial structures. Nature provides several protein nanoparticle platforms, including viral capsids, ferritin, and albumin, which hold significant potential for disease treatment. These naturally occurring protein nanoparticles not only serve as effective drug delivery platforms but also augment antigen delivery and targeting capabilities through techniques like genetic modification and covalent conjugation. Motivated by nature's originality and driven by progress in computational methodologies, scientists have crafted numerous protein nanoparticles with intricate assembly structures, showing significant potential in the development of multivalent vaccines. Consequently, both naturally occurring and de novo designed protein nanoparticles are anticipated to enhance the effectiveness of immunotherapy. This review consolidates the advancements in protein nanoparticles for immunotherapy across diseases including cancer and other diseases like influenza, pneumonia, and hepatitis.
Collapse
Affiliation(s)
- Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Siyuan Wei
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Lifan Duan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Yifan Ji
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xiaofan Han
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Qipeng Sun
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
27
|
Garmeh Motlagh F, Azimzadeh Irani M, Masoomi Nomandan SZ, Assadizadeh M. Computational design and investigation of the monomeric spike SARS-CoV-2-ferritin nanocage vaccine stability and interactions. Front Mol Biosci 2024; 11:1403635. [PMID: 38933369 PMCID: PMC11199398 DOI: 10.3389/fmolb.2024.1403635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Since the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) outbreak, several solutions have been proposed to manage the disease. The most viable option for controlling this virus is to produce effective vaccines. Most of the current SARS-CoV-2 vaccines have focused on the infusion spike protein. Spike exists as a trimer and plays a vital role in infecting host cells by binding to the Angiotensin-Converting Enzyme 2 (ACE2) receptor through its Receptor Binding Domain (RBD). Ferritin protein, a naturally occurring iron-storage protein, has gained attention for vaccine production due to its self-assembling property, non-toxic nature, and biocompatibility. Ferritin nanocages have recently been employed in the development of a SARS-CoV-2 vaccination eliciting not only long-term protective memory cells but also a sustained antibody response. In this study, a combination of in silico investigations including molecular docking, molecular dynamics simulations, and immune simulations were carried out to computationally model the monomeric spike protein on the ferritin nanocage as well as to evaluate its stability and interactions for the first time. The structural dynamics of the modeled complex demonstrated noticeable stability. In particular, the Receptor Binding Domain (RBD) and ferritin within the monomeric spike-ferritin complex illustrated significant stability. The lack of alterations in the secondary structure further supported the overall steadiness of the complex. The decline in the distance between ferritin and spike suggests a strong interaction over time. The cross-correlation matrices revealed that the monomeric spike and ferritin move towards each other supporting the stable interaction between spike and ferritin. Further, the orientation of monomeric spike protein within the ferritin unit facilitated the exposure of critical epitopes, specifically upward active Receptor Binding Domain (RBD), enabling effective interactions with the ACE2 receptor. The immune simulations of the model indicated high-level stimulations of both cellular and humoral immunity in the human body. It was also found that the employed model is effective regardless of the mutated spikes in different variants. These findings shed light on the current status of the SARS-CoV-2-ferritin nanoparticle vaccines and could be used as a framework for other similar vaccine designs.
Collapse
|
28
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-z] [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: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
29
|
Yu Y, Shi Y, Kwon YW, Choi Y, Kim Y, Na JG, Huh J, Lee J. A rationally designed miniature of soluble methane monooxygenase enables rapid and high-yield methanol production in Escherichia coli. Nat Commun 2024; 15:4399. [PMID: 38782897 PMCID: PMC11116448 DOI: 10.1038/s41467-024-48671-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Soluble methane monooxygenase (sMMO) oxidizes a wide range of carbon feedstocks (C1 to C8) directly using intracellular NADH and is a useful means in developing green routes for industrial manufacturing of chemicals. However, the high-throughput biosynthesis of active recombinant sMMO and the ensuing catalytic oxidation have so far been unsuccessful due to the structural and functional complexity of sMMO, comprised of three functionally complementary components, which remains a major challenge for its industrial applications. Here we develop a catalytically active miniature of sMMO (mini-sMMO), with a turnover frequency of 0.32 s-1, through an optimal reassembly of minimal and modified components of sMMO on catalytically inert and stable apoferritin scaffold. We characterise the molecular characteristics in detail through in silico and experimental analyses and verifications. Notably, in-situ methanol production in a high-cell-density culture of mini-sMMO-expressing recombinant Escherichia coli resulted in higher yield and productivity (~ 3.0 g/L and 0.11 g/L/h, respectively) compared to traditional methanotrophic production.
Collapse
Affiliation(s)
- Yeonhwa Yu
- Department of Chemical and Biological Engineering, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea
| | - Yongfan Shi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Young Wan Kwon
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea
| | - Yoobin Choi
- Department of Chemical and Biological Engineering, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea
| | - Yusik Kim
- Department of Chemical and Biological Engineering, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea
| | - Jeong-Geol Na
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - June Huh
- Department of Chemical and Biological Engineering, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea.
| | - Jeewon Lee
- Department of Chemical and Biological Engineering, Korea University, Anam-Dong 5-1, Seongbuk-Gu, Seoul, 02841, Republic of Korea.
| |
Collapse
|
30
|
Guan W, Gao H, Liu Y, Sun S, Li G. Application of magnetism in tissue regeneration: recent progress and future prospects. Regen Biomater 2024; 11:rbae048. [PMID: 38939044 PMCID: PMC11208728 DOI: 10.1093/rb/rbae048] [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: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 06/29/2024] Open
Abstract
Tissue regeneration is a hot topic in the field of biomedical research in this century. Material composition, surface topology, light, ultrasonic, electric field and magnetic fields (MFs) all have important effects on the regeneration process. Among them, MFs can provide nearly non-invasive signal transmission within biological tissues, and magnetic materials can convert MFs into a series of signals related to biological processes, such as mechanical force, magnetic heat, drug release, etc. By adjusting the MFs and magnetic materials, desired cellular or molecular-level responses can be achieved to promote better tissue regeneration. This review summarizes the definition, classification and latest progress of MFs and magnetic materials in tissue engineering. It also explores the differences and potential applications of MFs in different tissue cells, aiming to connect the applications of magnetism in various subfields of tissue engineering and provide new insights for the use of magnetism in tissue regeneration.
Collapse
Affiliation(s)
- Wenchao Guan
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Hongxia Gao
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yaqiong Liu
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Shaolan Sun
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
31
|
Kwon S, Andreas MP, Giessen TW. Pore engineering as a general strategy to improve protein-based enzyme nanoreactor performance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592161. [PMID: 38746127 PMCID: PMC11092584 DOI: 10.1101/2024.05.02.592161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Enzyme nanoreactors are nanoscale compartments consisting of encapsulated enzymes and a selectively permeable barrier. Sequestration and co-localization of enzymes can increase catalytic activity, stability, and longevity, highly desirable features for many biotechnological and biomedical applications of enzyme catalysts. One promising strategy to construct enzyme nanoreactors is to repurpose protein nanocages found in nature. However, protein-based enzyme nanoreactors often exhibit decreased catalytic activity, partially caused by a mismatch of protein shell selectivity and the substrate requirements of encapsulated enzymes. No broadly applicable and modular protein-based nanoreactor platform is currently available. Here, we introduce a pore-engineered universal enzyme nanoreactor platform based on encapsulins - microbial self-assembling protein nanocompartments with programmable and selective enzyme packaging capabilities. We structurally characterize our protein shell designs via cryo-electron microscopy and highlight their polymorphic nature. Through fluorescence polarization assays, we show their improved molecular flux behavior and highlight their expanded substrate range via a number of proof-of-concept enzyme nanoreactor designs. This work lays the foundation for utilizing our encapsulin-based nanoreactor platform for future biotechnological and biomedical applications.
Collapse
Affiliation(s)
- Seokmu Kwon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michael P. Andreas
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tobias W. Giessen
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Hori M, Steinauer A, Tetter S, Hälg J, Manz EM, Hilvert D. Stimulus-responsive assembly of nonviral nucleocapsids. Nat Commun 2024; 15:3576. [PMID: 38678040 PMCID: PMC11055949 DOI: 10.1038/s41467-024-47808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
Controlled assembly of a protein shell around a viral genome is a key step in the life cycle of many viruses. Here we report a strategy for regulating the co-assembly of nonviral proteins and nucleic acids into highly ordered nucleocapsids in vitro. By fusing maltose binding protein to the subunits of NC-4, an engineered protein cage that encapsulates its own encoding mRNA, we successfully blocked spontaneous capsid assembly, allowing isolation of the individual monomers in soluble form. To initiate RNA-templated nucleocapsid formation, the steric block can be simply removed by selective proteolysis. Analyses by transmission and cryo-electron microscopy confirmed that the resulting assemblies are structurally identical to their RNA-containing counterparts produced in vivo. Enzymatically triggered cage formation broadens the range of RNA molecules that can be encapsulated by NC-4, provides unique opportunities to study the co-assembly of capsid and cargo, and could be useful for studying other nonviral and viral assemblies.
Collapse
Affiliation(s)
- Mao Hori
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, Japan
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LIBN, Lausanne, Switzerland
| | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| | - Jamiro Hälg
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland
| | - Eva-Maria Manz
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
33
|
Padariya M, Kalathiya U. Single Ferritin Nanocages Expressing SARS-CoV-2 Spike Variants to Receptor and Antibodies. Vaccines (Basel) 2024; 12:446. [PMID: 38793697 PMCID: PMC11125617 DOI: 10.3390/vaccines12050446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
SARS-CoV-2 virus variants of concern (VOCs) have rapidly changed their transmissibility and pathogenicity primarily through mutations in the structural proteins. Herein, we present molecular details with dynamics of the ferritin nanocages stitched with synthetic chimeras displaying the Spike receptor binding domains (RBDs). Our findings demonstrated the potential usage of ferritin-based vaccines that may effectively inhibit viral entry by blocking the Spike-ACE2 network and may induce cross-protective antibody responses. Taking the nanocage constructs into consideration, we evaluated the effects of variants on the docked interface of the SARS-CoV-2 Spike RBD with the ACE2 (angiotensin-converting enzyme 2) host cell receptor and neutralizing antibodies (Abs). Investigating the VOCs revealed that most of the mutations reported a possibly reduced structural stability within the Spike RBD domain. Point mutations have moderate or no effect for VVH-72, CR3022, and S309 Abs when bound with the Spike RBD, whereas a significant effect was observed for B38, CB6, and m396 over the surface of the H-ferritin nanocage. In addition to providing useful therapeutic approaches against COVID-19 (coronavirus disease 2019), these structural details can also be used to fight future coronavirus outbreaks.
Collapse
Affiliation(s)
- Monikaben Padariya
- International Centre for Cancer Vaccine Science, University of Gdansk, ul. Kładki 24, 80-822 Gdansk, Poland
| | - Umesh Kalathiya
- International Centre for Cancer Vaccine Science, University of Gdansk, ul. Kładki 24, 80-822 Gdansk, Poland
| |
Collapse
|
34
|
Kaur H, Garg M, Tomar D, Singh S, Jena KC. Role of tungsten disulfide quantum dots in specific protein-protein interactions at air-water interface. J Chem Phys 2024; 160:084705. [PMID: 38411235 DOI: 10.1063/5.0187563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024] Open
Abstract
The intriguing network of antibody-antigen (Ab-Ag) interactions is highly governed by environmental perturbations and the nature of biomolecular interaction. Protein-protein interactions (PPIs) have potential applications in developing protein-adsorption-based sensors and nano-scale materials. Therefore, characterizing PPIs in the presence of a nanomaterial at the molecular level becomes imperative. The present work involves the investigation of antiferritin-ferritin (Ab-Ag) protein interactions under the influence of tungsten disulfide quantum dots (WS2 QDs). Isothermal calorimetry and contact angle measurements validated the strong influence of WS2 QDs on Ab-Ag interactions. The interfacial signatures of nano-bio-interactions were evaluated using sum frequency generation vibration spectroscopy (SFG-VS) at the air-water interface. Our SFG results reveal a variation in the tilt angle of methyl groups by ∼12° ± 2° for the Ab-Ag system in the presence of WS2 QDs. The results illustrated an enhanced ordering of water molecules in the presence of QDs, which underpins the active role of interfacial water molecules during nano-bio-interactions. We have also witnessed a differential impact of QDs on Ab-Ag by raising the concentration of the Ab-Ag combination, which showcased an increased inter-molecular interaction among the Ab and Ag molecules and a minimal influence on the methyl tilt angle. These findings suggest the formation of stronger and ordered Ab-Ag complexes upon introducing WS2 QDs in the aqueous medium and signify the potentiality of WS2 QDs relevant to protein-based sensing assays.
Collapse
Affiliation(s)
- Harsharan Kaur
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Mayank Garg
- CSIR-Central Scientific Instruments Organisation (CSIR-CSIO), Chandigarh 160030, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepak Tomar
- Department of Physics, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Suman Singh
- CSIR-Central Scientific Instruments Organisation (CSIR-CSIO), Chandigarh 160030, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kailash C Jena
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
- Department of Physics, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
35
|
Hendricks AR, Cohen RS, McEwen GA, Tien T, Guilliams BF, Alspach A, Snow CD, Ackerson CJ. Laboratory Evolution of Metalloid Reductase Substrate Recognition and Nanoparticle Product Size. ACS Chem Biol 2024; 19:289-299. [PMID: 38295274 DOI: 10.1021/acschembio.3c00493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Glutathione reductase-like metalloid reductase (GRLMR) is an enzyme that reduces selenodiglutathione (GS-Se-SG), forming zerovalent Se nanoparticles (SeNPs). Error-prone polymerase chain reaction was used to create a library of ∼10,000 GRLMR variants. The library was expressed in BL21Escherichia coli in liquid culture with 50 mM of SeO32- present, under the hypothesis that the enzyme variants with improved GS-Se-SG reduction kinetics would emerge. The selection resulted in a GRLMR variant with two mutations. One of the mutations (D-E) lacks an obvious functional role, whereas the other mutation is L-H within 5 Å of the enzyme active site. This mutation places a second H residue within 5 Å of an active site dicysteine. This GRLMR variant was characterized for NADPH-dependent reduction of GS-Se-SG, GSSG, SeO32-, SeO42-, GS-Te-SG, and TeO32-. The evolved enzyme demonstrated enhanced reduction of SeO32- and gained the ability to reduce SeO42-. This variant is named selenium reductase (SeR) because of its emergent broad activity for a wide variety of Se substrates, whereas the parent enzyme was specific for GS-Se-SG. This study overall suggests that new biosynthetic routes are possible for inorganic nanomaterials using laboratory-directed evolution methods.
Collapse
Affiliation(s)
- Alexander R Hendricks
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
| | - Rachel S Cohen
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Gavin A McEwen
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
| | - Tony Tien
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Bradley F Guilliams
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
| | - Audrey Alspach
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
| | - Christopher D Snow
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Christopher J Ackerson
- Department of Chemistry, Colorado State University, Fort Collins, Colorado 80523-1872, United States
| |
Collapse
|
36
|
Xia H, Xu H, Wang J, Wang C, Chen R, Tao T, Xu S, Zhang J, Ma K, Wang J. Heat sensitive E-helix cut ferritin nanocages for facile and high-efficiency loading of doxorubicin. Int J Biol Macromol 2023; 253:126973. [PMID: 37729988 DOI: 10.1016/j.ijbiomac.2023.126973] [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: 06/05/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023]
Abstract
Ferritin possesses a stable and uniform cage structure, along with tumor-targeting properties and excellent biocompatibility, making it a promising drug delivery vehicle. However, the current ferritin drug loading strategy involves complex steps and harsh reaction conditions, resulting in low yield and recovery of drug loading, which limits the clinical application prospects of ferritin nanomedicine. In this study, we utilized the high-efficiency heat-sensitivity of the multiple channel switch structures of the E-helix-cut ferritin mutant (Ecut-HFn) and Cu2+ assistance to achieve high-efficiency loading of chemotherapeutic drugs in a one-step process at low temperatures. This method features mild reaction conditions (45 °C), high loading efficiency (about 110 doxorubicin (Dox) per Ecut-HFn), and improved protein and Dox recovery rates (with protein recovery rate around 94 % and Dox recovery rate reaching up to 45 %). The prepared ferritin-Dox particles (Ecut-HFn-Cu-Dox) exhibit a uniform size distribution, good stability, and retain the natural tumor targeting ability of ferritin. Overall, this temperature-controlled drug loading strategy utilizing heat-sensitivity ferritin mutants is energy-saving, environmentally friendly, efficient, and easy to operate, offering a new perspective for scaling up the industrial production of ferritin drug carriers.
Collapse
Affiliation(s)
- Haining Xia
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Huangtao Xu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China
| | - Jiarong Wang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China
| | - Changhao Wang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Ruiguo Chen
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China
| | - Tongxiang Tao
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Shuai Xu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China
| | - Jing Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Kun Ma
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China.
| | - Junfeng Wang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, PR China; University of Science and Technology of China, Hefei 230026, PR China; Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China.
| |
Collapse
|
37
|
Li N, Wang P, Xie Y, Wang B, Zhu C, Xue L, Han X, Gu N, Sun J. Expression of clMagR/clCry4 protein in mBMSCs provides T 2-contrast enhancement of MRI. Acta Biomater 2023; 172:309-320. [PMID: 37778484 DOI: 10.1016/j.actbio.2023.09.039] [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: 03/25/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Here, we propose for the first time the evaluation of magnetosensitive clMagR/clCry4 as a magnetic resonance imaging (MRI) reporter gene that imparts sensitivity to endogenous contrast in eukaryotic organisms. Using a lentiviral vector, we introduced clMagR/clCry4 into C57BL/6 mice-derived bone marrow mesenchymal stem cells (mBMSCs), which could specifically bind with iron, significantly affected MRI transverse relaxation, and generated readily detectable contrast without adverse effects in vivo. Specifically, clMagR/clCry4 makes mBMSCs beneficial for enhancing the sensitivity of MRI-R2 for iron-bearing granules, in which cells recruit exogenous iron and convert these stores into an MRI-detectable contrast; this is not achievable with control cells. Additionally, Prussian blue staining was performed together with ultrathin cell slices to provide direct evidence of natural iron-bearing granules being detectable on MRI. Hence, it was inferred that the sensitivity of MRI detection should be correlated with clMagR/clCry4 and exogenous iron. Taken together, the clMagR/clCry4 has great potential as an MRI reporter gene. STATEMENT OF SIGNIFICANCE: In this study, we propose the evaluation of magnetosensitive clMagR/clCry4 as an MRI reporter gene, imparting detection sensitivity to eukaryotic mBMSCs for endogenous contrast. At this point, the clMagR and clCry4 were located within the cytoplasm and possibly influence each other. The clMagR/clCry4 makes mBMSCs beneficial for enhancing the sensitivity of MRI-R2 for iron-bearing granules, in which protein could specifically bind with iron and convert these stores into MRI-detectable contrast; this is not achieved by control cells. The viewpoint was speculated that the clMagR/clCry4 and exogenous iron were complementary to each other. Additionally, Prussian blue staining was performed together with TEM observations to provide direct evidence that the iron-bearing granules were sensitive to MRI.
Collapse
Affiliation(s)
- Nuan Li
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Peng Wang
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China; Department of Sports Medicine and Adult Reconstructive Surgery, the Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210008, China
| | - Yuanyuan Xie
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210008, China
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University, Nanjing 210008, China
| | - Chenzhuo Zhu
- Southeast University-Monash University Joint Graduate School, Southeast University, Suzhou 215123, China
| | - Le Xue
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiaofeng Han
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ning Gu
- Medical School, Nanjing University, Nanjing 210093, China
| | - Jianfei Sun
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
38
|
Pham TT, Abe S, Date K, Hirata K, Suzuki T, Ueno T. Displaying a Protein Cage on a Protein Crystal by In-Cell Crystal Engineering. NANO LETTERS 2023; 23:10118-10125. [PMID: 37955329 DOI: 10.1021/acs.nanolett.3c02117] [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: 11/14/2023]
Abstract
The development of solid biomaterials has rapidly progressed in recent years in applications in bionanotechnology. The immobilization of proteins, such as enzymes, within protein crystals is being used to develop solid catalysts and functionalized materials. However, an efficient method for encapsulating protein assemblies has not yet been established. This work presents a novel approach to displaying protein cages onto a crystalline protein scaffold using in-cell protein crystal engineering. The polyhedra crystal (PhC) scaffold, which displays a ferritin cage, was produced by coexpression of polyhedrin monomer (PhM) and H1-ferritin (H1-Fr) monomer in Escherichia coli. The H1-tag is derived from the H1-helix of PhM. Our technique represents a unique strategy for immobilizing protein assemblies onto in-cell protein crystals and is expected to contribute to various applications in bionanotechnology.
Collapse
Affiliation(s)
- Thuc Toan Pham
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| | - Satoshi Abe
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| | - Koki Date
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| | - Kunio Hirata
- SR Life Science Instrumentation Unit, RIKEN/SPring-8 Center, 1-1-1, Kouto, Sayo-cho, Sayo-gun 679-5148, Hyogo, Japan
| | - Taiga Suzuki
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| | - Takafumi Ueno
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
- Living Systems Materialogy (LiSM) Research Group, International Research Frontiers Initiative (IRFI), Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
39
|
Yin YD, Chen FF, Hu J, Yang L, Song XT, Wu GR, Xu M, Gu ZY. Solid-State Nanopore Distinguishes Ferritin and Apo-Ferritin with Identical Exteriors through Amplified Flexibility at Single-Molecule Level. Anal Chem 2023; 95:16496-16504. [PMID: 37916987 DOI: 10.1021/acs.analchem.3c02041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Protein identification and discrimination at the single-molecule level are big challenges. Solid-state nanopores as a sensitive biosensor have been used for protein analysis, although it is difficult to discriminate proteins with similar structures in the traditional discrimination method based on the current blockage fraction. Here, we select ferritin and apo-ferritin as the model proteins that exhibit identical exterior and different interior structures and verify the practicability of their discrimination with flexibility features by the strategy of gradually decreasing the nanopore size. We show that the larger nanopore (relative to the protein size) has no obvious effect on discriminating two proteins. Then, the comparable-sized nanopore plays a key role in discriminating two proteins based on the dwell time and fraction distribution, and the conformational changes of both proteins are also studied with this nanopore. Finally, in the smaller nanopore, the protein molecules are trapped rather than translocated, where two proteins are obviously discriminated through the current fluctuation caused by the vibration of proteins. This strategy has potential in the discrimination of other important similar proteins.
Collapse
Affiliation(s)
- Yun-Dong Yin
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fang-Fang Chen
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jun Hu
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Lei Yang
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xi-Tong Song
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Guo-Rong Wu
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ming Xu
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Zhi-Yuan Gu
- Jiangsu Key Laboratory of Biofunctional Materials, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of New Power Batteries, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
40
|
Zhu H, Luo H, Chang R, Yang Y, Liu D, Ji Y, Qin H, Rong H, Yin J. Protein-based delivery systems for RNA delivery. J Control Release 2023; 363:253-274. [PMID: 37741460 DOI: 10.1016/j.jconrel.2023.09.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
RNA-based therapeutics have emerged as promising approaches to modulate gene expression and generate therapeutic proteins or antigens capable of inducing immune responses to treat a variety of diseases, such as infectious diseases, cancers, immunologic disorders, and genetic disorders. However, the efficient delivery of RNA molecules into cells poses significant challenges due to their large molecular weight, negative charge, and susceptibility to degradation by RNase enzymes. To overcome these obstacles, viral and non-viral vectors have been developed, including lipid nanoparticles, viral vectors, proteins, dendritic macromolecules, among others. Among these carriers, protein-based delivery systems have garnered considerable attention due to their potential to address specific issues associated with nanoparticle-based systems, such as liver accumulation and immunogenicity. This review provides an overview of currently marketed RNA drugs, underscores the significance of RNA delivery vector development, delineates the essential characteristics of an ideal RNA delivery vector, and introduces existing protein carriers for RNA delivery. By offering valuable insights, this review aims to serve as a reference for the future development of protein-based delivery vectors for RNA therapeutics.
Collapse
Affiliation(s)
- Haichao Zhu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Luo
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Ruilong Chang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Yang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Yue Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, No. 206, Sixian Street, Baiyun District, Guiyang City 550014, Guizhou Province, China.
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China.
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
41
|
Bossoni L, Labra-Muñoz JA, van der Zant HSJ, Čaluković V, Lefering A, Egli R, Huber M. In-depth magnetometry and EPR analysis of the spin structure of human-liver ferritin: from DC to 9 GHz. Phys Chem Chem Phys 2023; 25:27694-27717. [PMID: 37812236 PMCID: PMC10583656 DOI: 10.1039/d3cp01358h] [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: 03/25/2023] [Accepted: 08/05/2023] [Indexed: 10/10/2023]
Abstract
Ferritin, the major iron storage protein in organisms, stores iron in the form of iron oxyhydroxide most likely involving phosphorous as a constituent, the mineral form of which is not well understood. Therefore, the question of how the ca. 2000 iron atoms in the ferritin core are magnetically coupled is still largely open. The ferritin core, with a diameter of 5-8 nm, is encapsulated in a protein shell that also catalyzes the uptake of iron and protects the core from outside interactions. Neurodegenerative disease is associated with iron imbalance, generating specific interest in the magnetic properties of ferritin. Here we present 9 GHz continuous wave EPR and a comprehensive set of magnetometry techniques including isothermal remanent magnetization (IRM) and AC susceptibility to elucidate the magnetic properties of the core of human liver ferritin. For the analysis of the magnetometry data, a new microscopic model of the ferritin-core spin structure is derived, showing that magnetic moment is generated by surface-spin canting, rather than defects. The analysis explicitly includes the distribution of magnetic parameters, such as the distribution of the magnetic moment. This microscopic model explains some of the inconsistencies resulting from previous analysis approaches. The main findings are a mean magnetic moment of 337μB with a standard deviation of 0.947μB. In contrast to previous reports, only a relatively small contribution of paramagnetic and ferrimagnetic phases is found, in the order of maximally 3%. For EPR, the over 30 mT wide signal of the ferritin core is analyzed using the model of the giant spin system [Fittipaldi et al., Phys. Chem. Chem. Phys., 2016, 18, 3591-3597]. Two components are needed minimally, and the broadening of these components suggests a broad distribution of the magnetic resonance parameters, the zero-field splitting, D, and the spin quantum number, S. We compare parameters from EPR and magnetometry and find that EPR is particularly sensitive to the surface spins of the core, revealing the potential to use EPR as a diagnostic for surface-spin disorder.
Collapse
Affiliation(s)
- Lucia Bossoni
- C. J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, The Netherlands
| | - Jacqueline A Labra-Muñoz
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, 2300 RA Leiden, The Netherlands.
- Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, The Netherlands
| | - Herre S J van der Zant
- Kavli Institute of Nanoscience, Delft University of Technology, 2628 CJ Delft, The Netherlands
| | - Vera Čaluković
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, 2300 RA Leiden, The Netherlands.
| | - Anton Lefering
- RST-FAME, Delft University of Technology, Delft, The Netherlands
| | - Ramon Egli
- GeoSphere Austria, Department of Geophysics, Howe Warte 38, 1190 Vienna, Austria.
| | - Martina Huber
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, 2300 RA Leiden, The Netherlands.
| |
Collapse
|
42
|
Liang A, Zhou W, Zhang H, Zhang J, Zhang XE, Fang T, Li F. Effects of Individual Amino Acids on the Blood Circulation of Biosynthetic Protein Nanocages: Toward Guidance on Surface Engineering. Adv Healthc Mater 2023; 12:e2300502. [PMID: 37067183 DOI: 10.1002/adhm.202300502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/04/2023] [Indexed: 04/18/2023]
Abstract
Protein nanocages (PNCs) hold great promise for developing multifunctional nanomedicines. Long blood circulation is a key requirement of PNCs for most in vivo application scenarios. In addition to the classical PEGylation strategy, short peptides with a specific sequence screened via phage display are also very effective in prolonging the blood half-life (t1/2 ) of PNCs. However, there is a lack of knowledge on how individual amino acids affect the circulation of PNCs. Here the effects of the 20 proteinogenic amino acids in the form of an X3 or X5 tag (X represents an amino acid) are explored on the pharmacokinetics of PNCs, which lead to the formation of a heatmap illustrating the extent of t1/2 prolongation by each proteinogenic amino acid. Significantly, oligo-lysine and oligo-arginine can effectively prolong the t1/2 of strongly negatively charged PNCs through charge neutralization, while oligo-cysteine can also do so, but via a different mechanism, mediating the covalent binding of PNCs with plasma albumin as a stealth material. These findings are extendible and offer guidance for surface-engineering biosynthetic PNCs and other nanoparticles.
Collapse
Affiliation(s)
- Ao Liang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Juan Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian-En Zhang
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ti Fang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
43
|
Han K, Zhang Z, Tezcan FA. Spatially Patterned, Porous Protein Crystals as Multifunctional Materials. J Am Chem Soc 2023; 145:19932-19944. [PMID: 37642457 DOI: 10.1021/jacs.3c06348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
While the primary use of protein crystals has historically been in crystallographic structure determination, they have recently emerged as promising materials with many advantageous properties such as high porosity, biocompatibility, stability, structural and functional versatility, and genetic/chemical tailorability. Here, we report that the utility of protein crystals as functional materials can be further augmented through their spatial patterning and control of their morphologies. To this end, we took advantage of the chemically and kinetically controllable nature of ferritin self-assembly and constructed core-shell crystals with chemically distinct domains, tunable structural patterns, and morphologies. The spatial organization within ferritin crystals enabled the generation of patterned, multi-enzyme frameworks with cooperative catalytic behavior. We further exploited the differential growth kinetics of ferritin crystal facets to assemble Janus-type architectures with an anisotropic arrangement of chemically distinct domains. These examples represent a step toward using protein crystals as reaction vessels for complex multi-step reactions and broadening their utility as functional, solid-state materials. Our results demonstrate that morphology control and spatial patterning, which are key concepts in materials science and nanotechnology, can also be applied for engineering protein crystals.
Collapse
Affiliation(s)
- Kenneth Han
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Zhiyin Zhang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - F Akif Tezcan
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Materials Science and Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
44
|
Chu K, Ding Z, Zysman-Colman E. Materials for Electrochemiluminescence: TADF, Hydrogen-Bonding, and Aggregation- and Crystallization-Induced Emission Luminophores. Chemistry 2023; 29:e202301504. [PMID: 37344360 DOI: 10.1002/chem.202301504] [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: 05/12/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 06/23/2023]
Abstract
Electrochemiluminescence (ECL) is a rapidly growing discipline with many analytical applications from immunoassays to single-molecule detection. At the forefront of ECL research is materials chemistry, which looks at engineering new materials and compounds exhibiting enhanced ECL efficiencies compared to conventional fluorescent materials. In this review, we summarize recent molecular design strategies that lead to high efficiency ECL. In particular, we feature recent advances in the use of thermally activated delayed fluorescence (TADF) emitters to produce enhanced electrochemiluminescence. We also document how hydrogen bonding, aggregation, and crystallization can each be recruited in the design of materials showing enhanced electrochemiluminescence.
Collapse
Affiliation(s)
- Kenneth Chu
- Department of Chemistry, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Zhifeng Ding
- Department of Chemistry, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Eli Zysman-Colman
- Organic Semiconductor Centre, EaStCHEM School of Chemistry, University of St Andrews, Fife, KY16 9ST, UK
| |
Collapse
|
45
|
Kumari A, Roy A. Enhancing micronutrient absorption through simultaneous fortification and phytic acid degradation. Food Sci Biotechnol 2023; 32:1235-1256. [PMID: 37362807 PMCID: PMC10290024 DOI: 10.1007/s10068-023-01255-8] [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: 10/28/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Abstract
Phytic acid (PA), an endogenous antinutrient in cereals and legumes, hinders mineral absorption by forming less bioavailable, stable PA-mineral complexes. For individual micronutrients, the PA-to-mineral molar ratio below the critical level ensures better bioavailability and is achieved by adding minerals or removing PA from cereals and pulses. Although several PA reduction and fortification strategies are available, the inability to completely eradicate or degrade PA using available techniques always subdues fortification's impact by hindering fortified micronutrient absorption. The bioavailability of micronutrients could be increased through simultaneous PA degradation and fortification. Following primary PA reduction of the raw material, the fortification step should also incorporate additional essential control stages to further PA inactivation, improving micronutrient absorption. In this review, the chemistry of PA interaction with metal ions, associated controlling parameters, and its impact on PA reduction during fortification is also evaluated, and further suggestions were made for the fortification's success.
Collapse
Affiliation(s)
- Ankanksha Kumari
- Laboratory of Applied Food Chemistry, Microbiology, and Process Engineering, Department of Chemical Engineering, Birla Institute of Technology Mesra, Ranchi, Jharkhand India
| | - Anupam Roy
- Laboratory of Applied Food Chemistry, Microbiology, and Process Engineering, Department of Chemical Engineering, Birla Institute of Technology Mesra, Ranchi, Jharkhand India
| |
Collapse
|
46
|
Yuan F, Su B, Yu Y, Wang J. Study and design of amino acid-based radical enzymes using unnatural amino acids. RSC Chem Biol 2023; 4:431-446. [PMID: 37292061 PMCID: PMC10246556 DOI: 10.1039/d2cb00250g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 05/17/2023] [Indexed: 06/10/2023] Open
Abstract
Radical enzymes harness the power of reactive radical species by placing them in a protein scaffold, and they are capable of catalysing many important reactions. New native radical enzymes, especially those with amino acid-based radicals, in the category of non-heme iron enzymes (including ribonucleotide reductases), heme enzymes, copper enzymes, and FAD-radical enzymes have been discovered and characterized. We discussed recent research efforts to discover new native amino acid-based radical enzymes, and to study the roles of radicals in processes such as enzyme catalysis and electron transfer. Furthermore, design of radical enzymes in a small and simple scaffold not only allows us to study the radical in a well-controlled system and test our understanding of the native enzymes, but also allows us to create powerful enzymes. In the study and design of amino acid-based radical enzymes, the use of unnatural amino acids allows precise control of pKa values and reduction potentials of the residue, as well as probing the location of the radical through spectroscopic methods, making it a powerful research tool. Our understanding of amino acid-based radical enzymes will allow us to tailor them to create powerful catalysts and better therapeutics.
Collapse
Affiliation(s)
- Feiyan Yuan
- Institute of Biochemical Engineering, Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 102488 China
| | - Binbin Su
- Institute of Biochemical Engineering, Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 102488 China
| | - Yang Yu
- Institute of Biochemical Engineering, Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology Beijing 102488 China
| | - Jiangyun Wang
- Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences Beijing 100101 China
| |
Collapse
|
47
|
Wang Y, Xiong Y, Duan Y, Shi K, Su C, Ding L, Wang J, He L. Activatable Fluorescence-Encoded Nanoprobes Enable Simple Multiplexed RNA Imaging in Live Cells. ACS Sens 2023; 8:1918-1928. [PMID: 37130214 DOI: 10.1021/acssensors.2c02657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Benefiting from superior programmable performance and flexible design of DNA technologies, a variety of single-molecule RNA fluorescence imaging methodologies have been reported. However, the multiplexing capability is restricted owing to the spectral overlap of fluorophores. To overcome this limitation, some inspiring multiplex imaging strategies have been developed, but in practice, it remains challenging to achieve convenient and rapid imaging in live cells due to complex designs and additional pretreatments to increase cell permeability. Here, we report an activatable fluorescence-encoded nanoprobe (AFENP) strategy, through which fluorescence-encoded functional modules for qualitative analysis and activated nucleic acid assemblies functional modules for quantitative testing enable simple multiplexed RNA imaging in single live cells. As a proof of principle, by two distinguishable fluorophores (fluorescein and rhodamine B) and their seven distinctly differentiated intensity levels, self-assembled AFENP enables simplified and quick simultaneous in situ detection and imaging of seven types of targets in live single cells because the fluorescent quantitative signal is activated only in the presence of target avoiding the washing procedures and additional pretreatment to increase cell permeability is undesired. We expect that this practical single-cell analysis platform will be adopted for multiple gene expression analysis and imaging in live cells on account of its simplicity and multiplex capability.
Collapse
Affiliation(s)
- Ya Wang
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Yamin Xiong
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Yanjuan Duan
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Kangqi Shi
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Chaojie Su
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Jia Wang
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Leiliang He
- College of Public Health, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| |
Collapse
|
48
|
Budiarta M, Roy S, Katenkamp T, Feliu N, Beck T. Overcoming Non-Specific Interactions for Efficient Encapsulation of Doxorubicin in Ferritin Nanocages for Targeted Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205606. [PMID: 36748864 DOI: 10.1002/smll.202205606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/22/2022] [Indexed: 05/25/2023]
Abstract
Due to its beneficial pharmacological properties, ferritin (Ftn) is considered as an interesting drug delivery vehicle to alleviate the cardiotoxicity of doxorubicin (DOX) in chemotherapy. However, the encapsulation of DOX in Ftn suffers from heavy precipitation and low protein recovery yield which limits its full potential. Here, a new DOX encapsulation strategy by cysteine-maleimide conjugation is proposed. In order to demonstrate that this strategy is more efficient compared to the other approaches, DOX is encapsulated in Ftn variants carrying different surface charges. Furthermore, in contrast to the common belief, this data show that DOX molecules are also found to bind non-specifically to the surface of Ftn. This can be circumvented by the use of Tris(2-carboxyethyl)phosphine (TCEP) during encapsulation or by washing with acidic buffer. The biocompatibility studies of the resulting DOX Ftn variants in MCF-7 and MHS cancer cells shows a complex relationship between the cytotoxicity, the DOX loading and the different surface charges of Ftn. Further investigation on the cell uptake mechanism provides reasonable explanations for the cytotoxicity results and reveals that surface charging of Ftn hinders its transferrin receptor 1 (TfR-1) mediated cellular uptake in MCF-7 cells.
Collapse
Affiliation(s)
- Made Budiarta
- Institute of Inorganic Chemistry, RWTH Aachen University, Landoltweg 1, 52074, Aachen, Germany
| | - Sathi Roy
- Fraunhofer Center for Applied Nanotechnology (CAN), Fraunhofer IAP, Grindelallee 117, 20146, Hamburg, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607, Hamburg, Germany
| | - Tobias Katenkamp
- Institute of Physical Chemistry, Department of Chemistry, Universität Hamburg, Grindelallee 117, 20146, Hamburg, Germany
| | - Neus Feliu
- Fraunhofer Center for Applied Nanotechnology (CAN), Fraunhofer IAP, Grindelallee 117, 20146, Hamburg, Germany
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22607, Hamburg, Germany
| | - Tobias Beck
- Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761, Hamburg, Germany
| |
Collapse
|
49
|
Yu J, Jin B, Ji Q, Wang H. Detoxification and metabolism of glyphosate by a Pseudomonas sp. via biogenic manganese oxidation. JOURNAL OF HAZARDOUS MATERIALS 2023; 448:130902. [PMID: 36731313 DOI: 10.1016/j.jhazmat.2023.130902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Biogenic manganese oxides (BMO) are widely distributed in groundwater and provides promise for adsorbing and oxidizing a wide range of micropollutants, however, the continuous biodegradation and bioavailability of micropollutants via cycle biogenic Mn(II) oxidation remains to be elucidated. In this study, glyphosate was degraded and to serve as the nutrient source by a Pseudomonas sp. QJX-1. The addition of glyphosate will not affect the Mn(II) oxidation function of the strain but will affect its Mn(II) oxidation process and effect. The glyphosate degradation products could further be used as the C, N and P sources for bacterium growth. Analysis of the RNA-seq data suggested that Mn(II) oxidation driven by oxidoreductases for glyphosate degradation. The long-term column experiments using biological Mn(II) cycling to realize continuous detoxification and metabolism of glyphosate, and thus revealed the synergism effects of biological and chemical conversion on toxic micropollutants and continuous metabolism in an aquatic ecosystem.
Collapse
Affiliation(s)
- Jie Yu
- Hebei Key Laboratory of Close-to-Nature Restoration Technology of Wetlands, School of Eco-Environment, Hebei University, Baoding 071002, China
| | - Boxuan Jin
- Hebei Key Laboratory of Close-to-Nature Restoration Technology of Wetlands, School of Eco-Environment, Hebei University, Baoding 071002, China
| | - Qinghua Ji
- Center for Water and Ecology, State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing 100084, China
| | - Hongjie Wang
- Hebei Key Laboratory of Close-to-Nature Restoration Technology of Wetlands, School of Eco-Environment, Hebei University, Baoding 071002, China; Institute of Xiong'an New Area, Hebei University, Baoding 071002, China; College of Life Science, Hebei University, Baoding 071002, China.
| |
Collapse
|
50
|
Qian W, Liu D, Han Y, Liu M, Liu B, Ji Q, Zhang B, Mei Q, Zhou S, Cheng Y. Cyclosporine A-loaded apoferritin alleviates myocardial ischemia-reperfusion injury by simultaneously blocking ferroptosis and apoptosis of cardiomyocytes. Acta Biomater 2023; 160:265-280. [PMID: 36822483 DOI: 10.1016/j.actbio.2023.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) seriously restricts the therapeutic effect of reperfusion. It is demonstrated that ferroptosis and apoptosis of cardiomyocytes are widely involved in MI/RI. Therefore, simultaneous inhibition of ferroptosis and apoptosis of cardiomyocytes can be a promising strategy to treat MI/RI. Besides, transferrin receptor 1 (TfR1) is highly expressed in ischemic myocardium, and apoferritin (ApoFn) is a ligand of the transferrin receptor. In this study, CsA@ApoFn was prepared by wrapping cyclosporin A (CsA) with ApoFn and actively accumulated in ischemic cardiomyocytes through TfR1 mediated endoctosis in MI/RI mice. After entering cardiomyocytes, ApoFn in CsA@ApoFn inhibited ferroptosis of ischemic cardiomyocytes by increasing the protein expression of GPX4 and reducing the content of labile iron pool and lipid peroxides. At the same time, CsA in CsA@ApoFn attenuated the apoptosis of ischemic cardiomyocytes through recovering mitochondrial membrane potential and reducing the level of reactive oxygen species, which played a synergistic role with ApoFn in the treatment of MI/RI. In conclusion, CsA@ApoFn restored cardiac function of MI/RI mice by simultaneously blocking ferroptosis and apoptosis of cardiomyocytes. ApoFn itself not only served as a safe carrier to specifically deliver CsA to ischemic cardiomyocytes but also played a therapeutic role on MI/RI. CsA@ApoFn is proved as an effective drug delivery platform for the treatment of MI/RI. STATEMENT OF SIGNIFICANCE: Recent studies have shown that ferroptosis is an important mechanism of myocardial ischemia-reperfusion injury (MI/RI). Therefore, simultaneous inhibition of ferroptosis and apoptosis of cardiomyocytes can be a promising strategy to treat MI/RI. Apoferritin, as a delivery carrier, can actively target to ischemic myocardium through binding with highly expressed transferrin receptor on ischemic cardiomyocytes. At the same time, apoferritin plays a protective role on ischemic cardiomyocytes by inhibiting ferroptosis. This strategy of killing two birds with one stone significantly improves the therapeutic effect on MI/RI while does not need more pharmaceutical excipients, which has the prospect of clinical transformation.
Collapse
Affiliation(s)
- Wenqiang Qian
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Ying Han
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Bao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Qibing Mei
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|