1
|
Georgiou E, Cabello-Garcia J, Xing Y, Howorka S. DNA Origami - Lipid Membrane Interactions Controlled by Nanoscale Sterics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404720. [PMID: 39162223 DOI: 10.1002/smll.202404720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/04/2024] [Indexed: 08/21/2024]
Abstract
DNA nanostructures designed to interact with bilayer membranes are of fundamental interest as they mimic biological cytoskeletons and other membrane-associated proteins for applications in synthetic biology, biosensing, and biological research. Yet, there is limited insight into how the binary interactions are influenced by steric effects produced by 3D geometries of DNA structures and membranes. This work uses a 3D DNA nanostructure with membrane anchors in four different steric environments to elucidate the interaction with membrane vesicles of varying sizes and different local bilayer morphology. It is found that interactions are significantly affected by the steric environments of the anchors -often against predicted accessibility- as well as local nanoscale morphology of bilayers rather than on the usually considered global vesicle size. Furthermore, anchor-mediated bilayer interactions are co-controlled by weak contacts with non-lipidated DNA regions, as showcased by pioneering size discrimination between 50 and 200 nm vesicles. This study extends DNA nanotechnology to controlled bilayer interactions and can facilitate the design of nanodevices for vesicle-based diagnostics, biosensing, and protocells.
Collapse
Affiliation(s)
- Elena Georgiou
- Department of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1H 0AJ, UK
| | - Javier Cabello-Garcia
- Department of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1H 0AJ, UK
| | - Yongzheng Xing
- National Engineering Research Center for Colloidal Materials, Shandong University, Jinan, Shandong, 250100, China
| | - Stefan Howorka
- Department of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1H 0AJ, UK
| |
Collapse
|
2
|
Bennett ID, Burns JR, Ryadnov MG, Howorka S, Pyne ALB. Lipidated DNA Nanostructures Target and Rupture Bacterial Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2207585. [PMID: 38840451 DOI: 10.1002/smll.202207585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/01/2024] [Indexed: 06/07/2024]
Abstract
Chemistry has the power to endow supramolecular nanostructures with new biomedically relevant functions. Here it is reported that DNA nanostructures modified with cholesterol tags disrupt bacterial membranes to cause microbial cell death. The lipidated DNA nanostructures bind more readily to cholesterol-free bacterial membranes than to cholesterol-rich, eukaryotic membranes. These highly negatively charged, lipidated DNA nanostructures cause bacterial cell death by rupturing membranes. Strikingly, killing is mediated by clusters of barrel-shaped nanostructures that adhere to the membrane without the involvement of expected bilayer-puncturing barrels. These DNA nanomaterials may inspire the development of polymeric or small-molecule antibacterial agents that mimic the principles of selective binding and rupturing to help combat antimicrobial resistance.
Collapse
Affiliation(s)
- Isabel D Bennett
- London Centre for Nanotechnology, University College London, 17-19 Gordon Street, London, WC1H 0AH, United Kingdom
- Division of Medicine, University College London, Cruciform Building, Gower Street, London, WC1E 6BT, United Kingdom
| | - Jonathan R Burns
- Department of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1H 0AJ, United Kingdom
| | - Maxim G Ryadnov
- National Physical Laboratory, Teddington, TW11 0LW, United Kingdom
- Department of Physics, King's College London, Strand Lane, London, WC2R 2LS, United Kingdom
| | - Stefan Howorka
- Department of Chemistry, Institute of Structural Molecular Biology, University College London, London, WC1H 0AJ, United Kingdom
| | - Alice L B Pyne
- London Centre for Nanotechnology, University College London, 17-19 Gordon Street, London, WC1H 0AH, United Kingdom
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Sheffield, S1 3JD, United Kingdom
| |
Collapse
|
3
|
Wang C, Lan X, Zhu L, Wang Y, Gao X, Li J, Tian H, Liang Z, Xu W. Construction Strategy of Functionalized Liposomes and Multidimensional Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309031. [PMID: 38258399 DOI: 10.1002/smll.202309031] [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: 10/08/2023] [Revised: 12/30/2023] [Indexed: 01/24/2024]
Abstract
Liposomes are widely used in the biological field due to their good biocompatibility and surface modification properties. With the development of biochemistry and material science, many liposome structures and their surface functional components have been modified and optimized one by one, pushing the liposome platform from traditional to functionalized and intelligent, which will better satisfy and expand the needs of scientific research. However, a main limiting factor effecting the efficiency of liposomes is the complicated environmental conditions in the living body. Currently, in order to overcome the above problem, functionalized liposomes have become a very promising strategy. In this paper, binding strategies of liposomes with four main functional elements, namely nucleic acids, antibodies, peptides, and stimuli-responsive motif have been summarized for the first time. In addition, based on the construction characteristics of functionalized liposomes, such as drug-carrying, targeting, long-circulating, and stimulus-responsive properties, a comprehensive overview of their features and respective research progress are presented. Finally, the paper critically presents the limitations of these functionalized liposomes in the current applications and also prospectively suggests the future development directions, aiming to accelerate realization of their industrialization.
Collapse
Affiliation(s)
- Chengyun Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, 071000, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Xinyue Lan
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Longjiao Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Yanhui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Xinru Gao
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
| | - Jie Li
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Hongtao Tian
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, 071000, China
| | - Zhihong Liang
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
| | - Wentao Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| |
Collapse
|
4
|
Samanta A, Baranda Pellejero L, Masukawa M, Walther A. DNA-empowered synthetic cells as minimalistic life forms. Nat Rev Chem 2024; 8:454-470. [PMID: 38750171 DOI: 10.1038/s41570-024-00606-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2024] [Indexed: 06/13/2024]
Abstract
Cells, the fundamental units of life, orchestrate intricate functions - motility, adaptation, replication, communication, and self-organization within tissues. Originating from spatiotemporally organized structures and machinery, coupled with information processing in signalling networks, cells embody the 'sensor-processor-actuator' paradigm. Can we glean insights from these processes to construct primitive artificial systems with life-like properties? Using de novo design approaches, what can we uncover about the evolutionary path of life? This Review discusses the strides made in crafting synthetic cells, utilizing the powerful toolbox of structural and dynamic DNA nanoscience. We describe how DNA can serve as a versatile tool for engineering entire synthetic cells or subcellular entities, and how DNA enables complex behaviour, including motility and information processing for adaptive and interactive processes. We chart future directions for DNA-empowered synthetic cells, envisioning interactive systems wherein synthetic cells communicate within communities and with living cells.
Collapse
Affiliation(s)
- Avik Samanta
- Life-Like Materials and Systems, Department of Chemistry, University of Mainz, Mainz, Germany.
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, India.
| | | | - Marcos Masukawa
- Life-Like Materials and Systems, Department of Chemistry, University of Mainz, Mainz, Germany
| | - Andreas Walther
- Life-Like Materials and Systems, Department of Chemistry, University of Mainz, Mainz, Germany.
| |
Collapse
|
5
|
Postigo A, Martínez-Vicente P, Baumann KN, Del Barrio J, Hernández-Ainsa S. Assessing the influence of small structural modifications in simple DNA-based nanostructures on their role as drug nanocarriers. Biomater Sci 2024; 12:1549-1557. [PMID: 38305143 DOI: 10.1039/d3bm01987j] [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: 02/03/2024]
Abstract
DNA nanotechnology leverages Watson-Crick-Franklin base-pairing interactions to build complex DNA-based nanostructures (DNS). Due to DNA specific self-assembly properties, DNS can be designed with a total control of their architecture, which has been demonstrated to have an impact on the overall DNS features. Indeed, structural properties such as the shape, size and flexibility of DNS can influence their biostability as well as their ability to internalise into cells. We present here two series of simple DNS with small and precise variations related to their length or flexibility and study the influence that these structural changes have on their overall properties as drug nanocarriers. Results indicate that shorter and more flexible DNS present higher stability towards nuclease degradation. These structural changes also have a certain effect on their cell internalisation ability and drug release rate. Consequently, drug-loaded DNS cytotoxicity varies according to the design, with lower cell viability values obtained in the DNS exhibiting faster drug release and larger cell interaction rates. In summary, small changes in the structure of simple DNS can have an influence on their overall capabilities as drug nanocarriers. The effects reported here could guide the design of simple DNS for future therapeutic uses.
Collapse
Affiliation(s)
- Alejandro Postigo
- Instituto de Nanociencia y Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Pablo Martínez-Vicente
- Instituto de Nanociencia y Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | | | - Jesús Del Barrio
- Instituto de Nanociencia y Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Silvia Hernández-Ainsa
- Instituto de Nanociencia y Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- ARAID Foundation, Government of Aragon, Zaragoza 50018, Spain
| |
Collapse
|
6
|
Jahnke K, Göpfrich K. Engineering DNA-based cytoskeletons for synthetic cells. Interface Focus 2023; 13:20230028. [PMID: 37577007 PMCID: PMC10415745 DOI: 10.1098/rsfs.2023.0028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023] Open
Abstract
The development and bottom-up assembly of synthetic cells with a functional cytoskeleton sets a major milestone to understand cell mechanics and to develop man-made machines on the nano- and microscale. However, natural cytoskeletal components can be difficult to purify, deliberately engineer and reconstitute within synthetic cells which therefore limits the realization of multifaceted functions of modern cytoskeletons in synthetic cells. Here, we review recent progress in the development of synthetic cytoskeletons made from deoxyribonucleic acid (DNA) as a complementary strategy. In particular, we explore the capabilities and limitations of DNA cytoskeletons to mimic functions of natural cystoskeletons like reversible assembly, cargo transport, force generation, mechanical support and guided polymerization. With recent examples, we showcase the power of rationally designed DNA cytoskeletons for bottom-up assembled synthetic cells as fully engineerable entities. Nevertheless, the realization of dynamic instability, self-replication and genetic encoding as well as contractile force generating motors remains a fruitful challenge for the complete integration of multifunctional DNA-based cytoskeletons into synthetic cells.
Collapse
Affiliation(s)
- Kevin Jahnke
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, 69120 Heidelberg, Germany
| | - Kerstin Göpfrich
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Center for Molecular Biology (ZMBH), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| |
Collapse
|
7
|
Yan Z, Liu Y, Zhao L, Hu J, Du Y, Peng X, Liu Z. In situ stimulus-responsive self-assembled nanomaterials for drug delivery and disease treatment. MATERIALS HORIZONS 2023; 10:3197-3217. [PMID: 37376926 DOI: 10.1039/d3mh00592e] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The individual motifs that respond to specific stimuli for the self-assembly of nanomaterials play important roles. In situ constructed nanomaterials are formed spontaneously without human intervention and have promising applications in bioscience. However, due to the complex physiological environment of the human body, designing stimulus-responsive self-assembled nanomaterials in vivo is a challenging problem for researchers. In this article, we discuss the self-assembly principles of various nanomaterials in response to the tissue microenvironment, cell membrane, and intracellular stimuli. We propose the applications and advantages of in situ self-assembly in drug delivery and disease diagnosis and treatment, with a focus on in situ self-assembly at the lesion site, especially in cancer. Additionally, we introduce the significance of introducing exogenous stimulation to construct self-assembly in vivo. Based on this foundation, we put forward the prospects and possible challenges in the field of in situ self-assembly. This review uncovers the relationship between the structure and properties of in situ self-assembled nanomaterials and provides new ideas for innovative drug molecular design and development to solve the problems in the targeted delivery and precision medicine.
Collapse
Affiliation(s)
- Ziling Yan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Licheng Zhao
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Jiaxin Hu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Yimin Du
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
- Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan Province, P. R. China
| |
Collapse
|
8
|
Yang J, Jahnke K, Xin L, Jing X, Zhan P, Peil A, Griffo A, Škugor M, Yang D, Fan S, Göpfrich K, Yan H, Wang P, Liu N. Modulating Lipid Membrane Morphology by Dynamic DNA Origami Networks. NANO LETTERS 2023. [PMID: 37440701 DOI: 10.1021/acs.nanolett.3c00750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
Membrane morphology and its dynamic adaptation regulate many cellular functions, which are often mediated by membrane proteins. Advances in DNA nanotechnology have enabled the realization of various protein-inspired structures and functions with precise control at the nanometer level, suggesting a viable tool to artificially engineer membrane morphology. In this work, we demonstrate a DNA origami cross (DOC) structure that can be anchored onto giant unilamellar vesicles (GUVs) and subsequently polymerized into micrometer-scale reconfigurable one-dimensional (1D) chains or two-dimensional (2D) lattices. Such DNA origami-based networks can be switched between left-handed (LH) and right-handed (RH) conformations by DNA fuels and exhibit potent efficacy in remodeling the membrane curvatures of GUVs. This work sheds light on designing hierarchically assembled dynamic DNA systems for the programmable modulation of synthetic cells for useful applications.
Collapse
Affiliation(s)
- Juanjuan Yang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, People's Republic of China
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Kevin Jahnke
- Biophysical Engineering Group, Max Planck Institute for Medical Research Heidelberg, Jahnstr. 29, 69120 Heidelberg, Germany
| | - Ling Xin
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Xinxin Jing
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Pengfei Zhan
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Andreas Peil
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Alessandra Griffo
- Biophysical Engineering Group, Max Planck Institute for Medical Research Heidelberg, Jahnstr. 29, 69120 Heidelberg, Germany
| | - Marko Škugor
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Donglei Yang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, People's Republic of China
| | - Sisi Fan
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
| | - Kerstin Göpfrich
- Biophysical Engineering Group, Max Planck Institute for Medical Research Heidelberg, Jahnstr. 29, 69120 Heidelberg, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), Im Neuenheime Feld 329, 69120 Heidelberg, Germany
| | - Hao Yan
- School of Molecular Sciences and Center for Molecular Design and Biomimetics at Biodesign Institute, Arizona State University, Tempe, Arizona 85287, United States
| | - Pengfei Wang
- Institute of Molecular Medicine, Department of Laboratory Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, People's Republic of China
| | - Na Liu
- 2. Physics Institute, University of Stuttgart, Pfaffenwaldring 57, 70569, Stuttgart, Germany
- Max Planck Institute for Solid State Research, Heisenbergstrasse 1, D-70569 Stuttgart, Germany
| |
Collapse
|
9
|
Zhan P, Peil A, Jiang Q, Wang D, Mousavi S, Xiong Q, Shen Q, Shang Y, Ding B, Lin C, Ke Y, Liu N. Recent Advances in DNA Origami-Engineered Nanomaterials and Applications. Chem Rev 2023; 123:3976-4050. [PMID: 36990451 PMCID: PMC10103138 DOI: 10.1021/acs.chemrev.3c00028] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Indexed: 03/31/2023]
Abstract
DNA nanotechnology is a unique field, where physics, chemistry, biology, mathematics, engineering, and materials science can elegantly converge. Since the original proposal of Nadrian Seeman, significant advances have been achieved in the past four decades. During this glory time, the DNA origami technique developed by Paul Rothemund further pushed the field forward with a vigorous momentum, fostering a plethora of concepts, models, methodologies, and applications that were not thought of before. This review focuses on the recent progress in DNA origami-engineered nanomaterials in the past five years, outlining the exciting achievements as well as the unexplored research avenues. We believe that the spirit and assets that Seeman left for scientists will continue to bring interdisciplinary innovations and useful applications to this field in the next decade.
Collapse
Affiliation(s)
- Pengfei Zhan
- 2nd Physics
Institute, University of Stuttgart, Pfaffenwaldring 57, 70569 Stuttgart, Germany
| | - Andreas Peil
- 2nd Physics
Institute, University of Stuttgart, Pfaffenwaldring 57, 70569 Stuttgart, Germany
| | - Qiao Jiang
- National
Center for Nanoscience and Technology, No 11, BeiYiTiao Zhongguancun, Beijing 100190, China
| | - Dongfang Wang
- School
of Biomedical Engineering and Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Shikufa Mousavi
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Qiancheng Xiong
- Department
of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Nanobiology
Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
| | - Qi Shen
- Department
of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Nanobiology
Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Department
of Molecular Biophysics and Biochemistry, Yale University, 266
Whitney Avenue, New Haven, Connecticut 06511, United States
| | - Yingxu Shang
- National
Center for Nanoscience and Technology, No 11, BeiYiTiao Zhongguancun, Beijing 100190, China
| | - Baoquan Ding
- National
Center for Nanoscience and Technology, No 11, BeiYiTiao Zhongguancun, Beijing 100190, China
| | - Chenxiang Lin
- Department
of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Nanobiology
Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Department
of Biomedical Engineering, Yale University, 17 Hillhouse Avenue, New Haven, Connecticut 06511, United States
| | - Yonggang Ke
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| | - Na Liu
- 2nd Physics
Institute, University of Stuttgart, Pfaffenwaldring 57, 70569 Stuttgart, Germany
- Max Planck
Institute for Solid State Research, Heisenbergstrasse 1, 70569 Stuttgart, Germany
| |
Collapse
|
10
|
Zhang Q, Gao L, Li F, Bi Y. Sensing and manipulating single lipid vesicles using dynamic DNA nanotechnology. NANOSCALE 2023; 15:5158-5166. [PMID: 36825547 DOI: 10.1039/d2nr07192d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Natural and artificial lipid vesicles have been widely involved in nano-delivery, bio-analysis and diagnosis. For sensing and manipulating single lipid vesicles, dynamic DNA reactions were constructed inside or on the surface of lipid vesicles. In this review, we interpreted various ways of integrating lipid vesicles and dynamic DNA nanotechnology by summarizing the latest reports in bio-analysis and biomimetic cell research.
Collapse
Affiliation(s)
- Qi Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University, Tai'An, Shandong, 271016, P. R. China.
- Key laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Sichuan, 610064, P. R. China.
| | - Lu Gao
- Key laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Sichuan, 610064, P. R. China.
| | - Feng Li
- Key laboratory of Green Chemistry & Technology of Ministry of Education, College of Chemistry, Sichuan University, Sichuan, 610064, P. R. China.
| | - Yanping Bi
- School of Pharmaceutical Sciences, Shandong First Medical University, Tai'An, Shandong, 271016, P. R. China.
| |
Collapse
|
11
|
Curcumin-Encapsulated Nanomicelles Improve Cellular Uptake and Cytotoxicity in Cisplatin-Resistant Human Oral Cancer Cells. J Funct Biomater 2022; 13:jfb13040158. [PMID: 36278627 PMCID: PMC9589971 DOI: 10.3390/jfb13040158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 12/24/2022] Open
Abstract
Oral cancer has a high mortality rate, which is mostly determined by the stage of the disease at the time of admission. Around half of all patients with oral cancer report with advanced illness. Hitherto, chemotherapy is preferred to treat oral cancer, but the emergence of resistance to anti-cancer drugs is likely to occur after a sequence of treatments. Curcumin is renowned for its anticancer potential but its marred water solubility and poor bioavailability limit its use in treating multidrug-resistant cancers. As part of this investigation, we prepared and characterized Curcumin nanomicelles (CUR-NMs) using DSPE-PEG-2000 and evaluated the anticancer properties of cisplatin-resistant cancer cell lines. The prepared CUR-NMs were sphere-shaped and unilamellar in structure, with a size of 32.60 ± 4.2 nm. CUR-NMs exhibited high entrapment efficiency (82.2%), entrapment content (147.96 µg/mL), and a mean zeta potential of −17.5ζ which is considered moderately stable. The cellular uptake and cytotoxicity studies revealed that CUR-NMs had significantly higher cytotoxicity and cellular uptake in cisplatin drug-resistant oral cancer cell lines and parental oral cancer cells compared to plain curcumin (CUR). The DAPI and FACS analysis corroborated a high percentage of apoptotic cells with CUR-NMs (31.14%) compared to neat CUR (19.72%) treatment. Conclusively, CUR-NMs can potentially be used as an alternative carrier system to improve the therapeutic effects of curcumin in the treatment of cisplatin-resistant human oral cancer.
Collapse
|
12
|
Ji X, Li Q, Song H, Fan C. Protein-Mimicking Nanoparticles in Biosystems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201562. [PMID: 35576606 DOI: 10.1002/adma.202201562] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Proteins are essential elements for almost all life activities. The emergence of nanotechnology offers innovative strategies to create a diversity of nanoparticles (NPs) with intrinsic capacities of mimicking the functions of proteins. These artificial mimics are produced in a cost-efficient and controllable manner, with their protein-mimicking performances comparable or superior to those of natural proteins. Moreover, they can be endowed with additional functionalities that are absent in natural proteins, such as cargo loading, active targeting, membrane penetrating, and multistimuli responding. Therefore, protein-mimicking NPs have been utilized more and more often in biosystems for a wide range of applications including detection, imaging, diagnosis, and therapy. To highlight recent progress in this broad field, herein, representative protein-mimicking NPs that fall into one of the four distinct categories are summarized: mimics of enzymes (nanozymes), mimics of fluorescent proteins, NPs with high affinity binding to specific proteins or DNA sequences, and mimics of protein scaffolds. This review covers their subclassifications, characteristic features, functioning mechanisms, as well as the extensive exploitation of their great potential for biological and biomedical purposes. Finally, the challenges and prospects in future development of protein-mimicking NPs are discussed.
Collapse
Affiliation(s)
- Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
13
|
Zhang H, Yu J, Ma L, Zhao Y, Xu S, Shi J, Qian K, Gu M, Tan H, Xu L, Liu Y, Mu C, Xiong Y. Reversing multi-drug resistance by polymeric metformin to enhance antitumor efficacy of chemotherapy. Int J Pharm 2022; 624:121931. [PMID: 35750278 DOI: 10.1016/j.ijpharm.2022.121931] [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: 03/19/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 01/01/2023]
Abstract
Multi-drug resistance (MDR) in breast cancer poses a great threat to chemotherapy. The expression and function of the ATP binding cassette (ABC) transporter are the major cause of MDR. Herein, a linear polyethylene glycol (PEI) conjugated with dicyandiamide, which called polymeric metformin (PolyMet), was successfully synthesized as a simple and biocompatible polymer of metformin. PolyMet showed the potential to reverse MDR by inhibiting the efflux of the substrate of ATP-binding cassette (ABC) transporter from DOX resistant MCF-7 cells (MCF-7/DOX). To test its MDR reversing effect, PolyMet was combined with DOX to treat mice carrying MCF-7/DOX xenografts. In order to decrease the toxicities of DOX and delivery PolyMet and DOX to tumor at the same time, PolyMet was complexed with poly-γ-glutamic acid-doxorubicin (PGA-DOX) electrostatically at the optimal ratio of 2:3, which were further coated with lipid membrane to form lipid/PolyMet-(PGA-DOX) nanoparticles (LPPD). The particle size of LPPD was 165.8 nm, and the zeta potential was +36.5 mV. LPPD exhibited favorable cytotoxicity and cellular uptake in MCF-7/DOX. Meanwhile, the bioluminescence imaging and immunohistochemical analysis indicated that LPPD effectively conquered DOX-associated MDR by blocking ABC transporters (ABCB1 and ABCC1) via PolyMet. Remarkably, LPPD significantly inhibited the tumor growth and lowered the systemic toxicity in a murine MCF-7/DOX tumor model. This is the first time to reveal that PolyMet can enhance the anti-tumor efficacy of DOX by dampening ABC transporters and activating the AMPK/mTOR pathway, which is a promising strategy for drug-resistant breast cancer therapy.
Collapse
Affiliation(s)
- Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiandong Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Lisha Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yue Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shujun Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ke Qian
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Mancang Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongsheng Tan
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Li Xu
- Zhejiang Provincial Hospital of TCM (Traditional Chinese Medicine), The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, China
| | - Yun Liu
- UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27559, USA
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
14
|
Baumann KN, Schröder T, Ciryam PS, Morzy D, Tinnefeld P, Knowles TPJ, Hernández-Ainsa S. DNA-Liposome Hybrid Carriers for Triggered Cargo Release. ACS APPLIED BIO MATERIALS 2022; 5:3713-3721. [PMID: 35838663 PMCID: PMC9382633 DOI: 10.1021/acsabm.2c00225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
The design of simple and versatile synthetic routes to
accomplish
triggered-release properties in carriers is of particular interest
for drug delivery purposes. In this context, the programmability and
adaptability of DNA nanoarchitectures in combination with liposomes
have great potential to render biocompatible hybrid carriers for triggered
cargo release. We present an approach to form a DNA mesh on large
unilamellar liposomes incorporating a stimuli-responsive DNA building
block. Upon incubation with a single-stranded DNA trigger sequence,
a hairpin closes, and the DNA building block is allowed to self-contract.
We demonstrate the actuation of this building block by single-molecule
Förster resonance energy transfer (FRET), fluorescence recovery
after photobleaching, and fluorescence quenching measurements. By
triggering this process, we demonstrate the elevated release of the
dye calcein from the DNA–liposome hybrid carriers. Interestingly,
the incubation of the doxorubicin-laden active hybrid carrier with
HEK293T cells suggests increased cytotoxicity relative to a control
carrier without the triggered-release mechanism. In the future, the
trigger could be provided by peritumoral nucleic acid sequences and
lead to site-selective release of encapsulated chemotherapeutics.
Collapse
Affiliation(s)
- Kevin N Baumann
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.,Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, U.K
| | - Tim Schröder
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 München, Germany
| | - Prashanth S Ciryam
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Diana Morzy
- Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, U.K
| | - Philip Tinnefeld
- Department of Chemistry and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 München, Germany
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.,Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, U.K
| | - Silvia Hernández-Ainsa
- Instituto de Nanociencia y Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.,Government of Aragon, ARAID Foundation, Zaragoza 50018, Spain
| |
Collapse
|
15
|
Nano-bio interactions: A major principle in the dynamic biological processes of nano-assemblies. Adv Drug Deliv Rev 2022; 186:114318. [PMID: 35533787 DOI: 10.1016/j.addr.2022.114318] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 12/18/2022]
Abstract
Controllable nano-assembly with stimuli-responsive groups is emerging as a powerful strategy to generate theranostic nanosystems that meet unique requirements in modern medicine. However, this prospective field is still in a proof-of-concept stage due to the gaps in our understanding of complex-(nano-assemblies)-complex-(biosystems) interactions. Indeed, stimuli-responsive assembly-disassembly is, in and of itself, a process of nano-bio interactions, the key steps for biological fate and functional activity of nano-assemblies. To provide a comprehensive understanding of these interactions in this review, we first propose a 4W1H principle (Where, When, What, Which and How) to delineate the relevant dynamic biological processes, behaviour and fate of nano-assemblies. We further summarize several key parameters that govern effective nano-bio interactions. The effects of these kinetic parameters on ADMET processes (absorption, distribution, metabolism, excretion and transformation) are then discussed. Furthermore, we provide an overview of the challenges facing the evaluation of nano-bio interactions of assembled nanodrugs. We finally conclude with future perspectives on safe-by-design and application-driven-design of nano-assemblies. This review will highlight the dynamic biological and physicochemical parameters of nano-bio interactions and bridge discrete concepts to build a full spectrum understanding of the biological outcomes of nano-assemblies. These principles are expected to pave the way for future development and clinical translation of precise, safe and effective nanomedicines with intelligent theranostic features.
Collapse
|
16
|
Lanphere C, Ciccone J, Dorey A, Hagleitner-Ertuğrul N, Knyazev D, Haider S, Howorka S. Triggered Assembly of a DNA-Based Membrane Channel. J Am Chem Soc 2022; 144:4333-4344. [PMID: 35253434 PMCID: PMC8931747 DOI: 10.1021/jacs.1c06598] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Indexed: 01/01/2023]
Abstract
Chemistry is in a powerful position to synthetically replicate biomolecular structures. Adding functional complexity is key to increase the biomimetics' value for science and technology yet is difficult to achieve with poorly controlled building materials. Here, we use defined DNA blocks to rationally design a triggerable synthetic nanopore that integrates multiple functions of biological membrane proteins. Soluble triggers bind via molecular recognition to the nanopore components changing their structure and membrane position, which controls the assembly into a defined channel for efficient transmembrane cargo transport. Using ensemble, single-molecule, and simulation analysis, our activatable pore provides insight into the kinetics and structural dynamics of DNA assembly at the membrane interface. The triggered channel advances functional DNA nanotechnology and synthetic biology and will guide the design of controlled nanodevices for sensing, cell biological research, and drug delivery.
Collapse
Affiliation(s)
- Conor Lanphere
- Department
of Chemistry, Institute of Structural Molecular Biology, University College London, London WC1H 0AJ, United Kingdom
| | - Jonah Ciccone
- Department
of Chemistry, Institute of Structural Molecular Biology, University College London, London WC1H 0AJ, United Kingdom
| | - Adam Dorey
- Department
of Chemistry, Institute of Structural Molecular Biology, University College London, London WC1H 0AJ, United Kingdom
| | | | - Denis Knyazev
- Institute
of Applied Experimental Biophysics, Johannes
Kepler University, 4040 Linz, Austria
| | - Shozeb Haider
- Department
of Pharmaceutical and Biological Chemistry, University College London School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Stefan Howorka
- Department
of Chemistry, Institute of Structural Molecular Biology, University College London, London WC1H 0AJ, United Kingdom
- Institute
of Applied Experimental Biophysics, Johannes
Kepler University, 4040 Linz, Austria
| |
Collapse
|
17
|
Amara U, Rashid S, Mahmood K, Nawaz MH, Hayat A, Hassan M. Insight into prognostics, diagnostics, and management strategies for SARS CoV-2. RSC Adv 2022; 12:8059-8094. [PMID: 35424750 PMCID: PMC8982343 DOI: 10.1039/d1ra07988c] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/04/2022] [Indexed: 01/08/2023] Open
Abstract
The foremost challenge in countering infectious diseases is the shortage of effective therapeutics. The emergence of coronavirus disease (COVID-19) outbreak has posed a great menace to the public health system globally, prompting unprecedented endeavors to contain the virus. Many countries have organized research programs for therapeutics and management development. However, the longstanding process has forced authorities to implement widespread infrastructures for detailed prognostic and diagnostics study of severe acute respiratory syndrome (SARS CoV-2). This review discussed nearly all the globally developed diagnostic methodologies reported for SARS CoV-2 detection. We have highlighted in detail the approaches for evaluating COVID-19 biomarkers along with the most employed nucleic acid- and protein-based detection methodologies and the causes of their severe downfall and rejection. As the variable variants of SARS CoV-2 came into the picture, we captured the breadth of newly integrated digital sensing prototypes comprised of plasmonic and field-effect transistor-based sensors along with commercially available food and drug administration (FDA) approved detection kits. However, more efforts are required to exploit the available resources to manufacture cheap and robust diagnostic methodologies. Likewise, the visualization and characterization tools along with the current challenges associated with waste-water surveillance, food security, contact tracing, and their role during this intense period of the pandemic have also been discussed. We expect that the integrated data will be supportive and aid in the evaluation of sensing technologies not only in current but also future pandemics.
Collapse
Affiliation(s)
- Umay Amara
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan 608000 Pakistan
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus 54000 Pakistan
| | - Sidra Rashid
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus 54000 Pakistan
| | - Khalid Mahmood
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan 608000 Pakistan
| | - Mian Hasnain Nawaz
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus 54000 Pakistan
| | - Akhtar Hayat
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus 54000 Pakistan
| | - Maria Hassan
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan 608000 Pakistan
| |
Collapse
|
18
|
Ma W, Yang Y, Zhu J, Jia W, Zhang T, Liu Z, Chen X, Lin Y. Biomimetic Nanoerythrosome-Coated Aptamer-DNA Tetrahedron/Maytansine Conjugates: pH-Responsive and Targeted Cytotoxicity for HER2-Positive Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109609. [PMID: 35064993 DOI: 10.1002/adma.202109609] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/19/2022] [Indexed: 02/05/2023]
Abstract
DNA materials have emerged as potential nanocarriers for targeted cancer therapy to precisely deliver cargos with specific purposes. The short half-life and low bioavailability of DNA materials due to their interception by the reticuloendothelial system and blood clearance further limit their clinical translation. This study employs an HER2-targeted DNA-aptamer-modified DNA tetrahedron (HApt-tFNA) as a drug delivery system, and combines maytansine (DM1) to develop the HApt-DNA tetrahedron/DM1 conjugate (HApt-tFNA@DM1, HTD, HApDC) for targeted therapy of HER2-positive cancer. To optimize the pharmacokinetics and tumor-aggregation of HTD, a biomimetic camouflage is applied to embed HTD. The biomimetic camouflage is constructed by merging the erythrocyte membrane with pH-responsive functionalized synthetic liposomes, thus with excellent performance of drug delivery and tumor-stimulated drug release. The hybrid erythrosome-based nanoparticles show better inhibition of HER2-positive cancer than other drug formulations and exhibit superior biosafety. With the strengths of precise delivery, increased drug loading, sensitive tumor probing, and prolonged circulation time, the HApDC represents a promising nanomedicine to treat HER2-positive tumors. Notably, this study developsa dual-targeting nanoparticle by combining pH-sensitive camouflage and HApDC, initiating an important step toward the development and application of DNA-based medicine and biomimetic cell membrane materials in cancer treatment and other potential biological applications.
Collapse
Affiliation(s)
- Wenjuan Ma
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Jianwei Zhu
- Department of Neurosurgery Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu 611731 P. R. China
| | - Weiqiang Jia
- Department of Neurosurgery Sichuan Provincial People's Hospital University of Electronic Science and Technology of China Chengdu 611731 P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Chengdu 610041 P. R. China
- College of Biomedical Engineering Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
19
|
Chen L, Zhang J, Lin Z, Zhang Z, Mao M, Wu J, Li Q, Zhang Y, Fan C. Pharmaceutical applications of framework nucleic acids. Acta Pharm Sin B 2022; 12:76-91. [PMID: 35127373 PMCID: PMC8799870 DOI: 10.1016/j.apsb.2021.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 01/21/2023] Open
Abstract
DNA is a biological polymer that encodes and stores genetic information in all living organism. Particularly, the precise nucleobase pairing inside DNA is exploited for the self-assembling of nanostructures with defined size, shape and functionality. These DNA nanostructures are known as framework nucleic acids (FNAs) for their skeleton-like features. Recently, FNAs have been explored in various fields ranging from physics, chemistry to biology. In this review, we mainly focus on the recent progress of FNAs in a pharmaceutical perspective. We summarize the advantages and applications of FNAs for drug discovery, drug delivery and drug analysis. We further discuss the drawbacks of FNAs and provide an outlook on the pharmaceutical research direction of FNAs in the future.
Collapse
Affiliation(s)
- Liang Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ziyan Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Miao Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiacheng Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
20
|
Zeng L, Wang H, Shi W, Chen L, Chen T, Chen G, Wang W, Lan J, Huang Z, Zhang J, Chen J. Aloe derived nanovesicle as a functional carrier for indocyanine green encapsulation and phototherapy. J Nanobiotechnology 2021; 19:439. [PMID: 34930289 PMCID: PMC8686546 DOI: 10.1186/s12951-021-01195-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/08/2021] [Indexed: 12/27/2022] Open
Abstract
Background Cancer is one of the devastating diseases in the world. The development of nanocarrier provides a promising perspective for improving cancer therapeutic efficacy. However, the issues with potential toxicity, quantity production, and excessive costs limit their further applications in clinical practice. Results Herein, we proposed a nanocarrier obtained from aloe with stability and leak-proofness. We isolated nanovesicles from the gel and rind of aloe (gADNVs and rADNVs) with higher quality and yield by controlling the final centrifugation time within 20 min, and modulating the viscosity at 2.98 mPa S and 1.57 mPa S respectively. The gADNVs showed great structure and storage stability, antioxidant and antidetergent capacity. They could be efficiently taken up by melanoma cells, and with no toxicity in vitro or in vivo. Indocyanine green (ICG) loaded in gADNVs (ICG/gADNVs) showed great stability in both heating system and in serum, and its retention rate exceeded 90% after 30 days stored in gADNVs. ICG/gADNVs stored 30 days could still effectively damage melanoma cells and inhibit melanoma growth, outperforming free ICG and ICG liposomes. Interestingly, gADNVs showed prominent penetrability to mice skin which might be beneficial to noninvasive transdermal administration. Conclusions Our research was designed to simplify the preparation of drug carrier, and reduce production cost, which provided an alternative for the development of economic and safe drug delivery system. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01195-7.
Collapse
Affiliation(s)
- Lupeng Zeng
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Huaying Wang
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Wanhua Shi
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Lingfan Chen
- Fujian Province New Drug Safety Evaluation Centre, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Tingting Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Guanyu Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Wenshen Wang
- Department of Chemical Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, People's Republic of China
| | - Jianming Lan
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Zhihong Huang
- Public Technology Service Center, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Jing Zhang
- Department of Chemical Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian, People's Republic of China.
| | - Jinghua Chen
- The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China. .,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.
| |
Collapse
|
21
|
Morzy D, Joshi H, Sandler SE, Aksimentiev A, Keyser UF. Membrane Activity of a DNA-Based Ion Channel Depends on the Stability of Its Double-Stranded Structure. NANO LETTERS 2021; 21:9789-9796. [PMID: 34767378 DOI: 10.1021/acs.nanolett.1c03791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
DNA nanotechnology has emerged as a promising method for designing spontaneously inserting and fully controllable synthetic ion channels. However, both insertion efficiency and stability of existing DNA-based membrane channels leave much room for improvement. Here, we demonstrate an approach to overcoming the unfavorable DNA-lipid interactions that hinder the formation of a stable transmembrane pore. Our all-atom MD simulations and experiments show that the insertion-driving cholesterol modifications can cause fraying of terminal base pairs of nicked DNA constructs, distorting them when embedded in a lipid bilayer. Importantly, we show that DNA nanostructures with no backbone discontinuities form more stable conductive pores and insert into membranes with a higher efficiency than the equivalent nicked constructs. Moreover, lack of nicks allows design and maintenance of membrane-spanning helices in a tilted orientation within the lipid bilayer. Thus, reducing the conformational degrees of freedom of the DNA nanostructures enables better control over their function as synthetic ion channels.
Collapse
Affiliation(s)
- Diana Morzy
- Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, United Kingdom
| | - Himanshu Joshi
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, Illinois 61801, United States
| | - Sarah E Sandler
- Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, United Kingdom
| | - Aleksei Aksimentiev
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana, Illinois 61801, United States
| | - Ulrich F Keyser
- Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, United Kingdom
| |
Collapse
|
22
|
Huang H, Belwal T, Li L, Xu Y, Zou L, Lin X, Luo Z. Amphiphilic and Biocompatible DNA Origami-Based Emulsion Formation and Nanopore Release for Anti-Melanogenesis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2104831. [PMID: 34608748 DOI: 10.1002/smll.202104831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/13/2021] [Indexed: 06/13/2023]
Abstract
Programmable engineered DNA origami provides infinite possibilities for customizing nanostructures with controllable precision and configurable functionality. Here, a strategy for fabricating an amphiphilic triangular DNA origami with a central nanopore that integrates phase-stabilizing, porous-gated, and affinity-delivering effects is presented. By introducing the DNA origami as a single-component surfactant, the water-in-oil-in-water (W/O/W) emulsion is effectively stabilized with decreased interfacial tension. Microscopic observation validates the attachment of the DNA origami onto the water-in-oil and oil-in-water interfaces. Furthermore, fluorescence studies and molecular docking simulations indicate the binding interactions of DNA origami with arbutin and coumaric acid at docking sites within central nanopores. These central nanopores are functionalized as molecular gates and affinity-based scaffold for the zero-order release of arbutin and coumaric acid at a constant rate regardless of concentration gradient throughout the whole releasing period. In vivo zebrafish results illustrate the advantages of this zero-order release for anti-melanogenesis therapy over direct exposure or Fickian diffusion. The DNA origami-based W/O/W emulsion presents anti-melanogenic effects against UV-B exposure without cardiotoxicity or motor toxicity. These results demonstrate that this non-toxic amphiphilic triangular DNA origami is capable of solely stabilizing the W/O/W emulsion as well as serving as nanopore gates and affinity-based scaffold for constant release.
Collapse
Affiliation(s)
- Hao Huang
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Tarun Belwal
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Li Li
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yanqun Xu
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Ligen Zou
- Hangzhou Academy of Agricultural Sciences, Hangzhou, 310024, China
| | - Xingyu Lin
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo, 315100, China
| | - Zisheng Luo
- Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- Ningbo Research Institute, Zhejiang University, Ningbo, 315100, China
| |
Collapse
|
23
|
He Q, Liu Y, Li K, Wu Y, Wang T, Tan Y, Jiang T, Liu X, Liu Z. Deoxyribonucleic acid anchored on cell membranes for biomedical application. Biomater Sci 2021; 9:6691-6717. [PMID: 34494042 DOI: 10.1039/d1bm01057c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Engineering cellular membranes with functional molecules provides an attractive strategy to manipulate cellular behaviors and functionalities. Currently, synthetic deoxyribonucleic acid (DNA) has emerged as a promising molecular tool to engineer cellular membranes for biomedical applications due to its molecular recognition and programmable properties. In this review, we summarized the recent advances in anchoring DNA on the cellular membranes and their applications. The strategies for anchoring DNA on cell membranes were summarized. Then their applications, such as immune response activation, receptor oligomerization regulation, membrane structure mimicking, cell-surface biosensing, and construction of cell clusters, were listed. The DNA-enabled intelligent systems which were able to sense stimuli such as DNA strands, light, and metal ions were highlighted. Finally, insights regarding the remaining challenges and possible future directions were provided.
Collapse
Affiliation(s)
- Qunye He
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ke Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yuwei Wu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yifu Tan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ting Jiang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Xiaoqin Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China. .,Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, P. R. China
| |
Collapse
|
24
|
Zhu L, Kuang Z, Song P, Li W, Gui L, Yang K, Ge F, Tao Y, Zhang W. Gold nanorod-loaded thermosensitive liposomes facilitate the targeted release of ruthenium(II) polypyridyl complexes with anti-tumor activity. NANOTECHNOLOGY 2021; 32:455103. [PMID: 34352731 DOI: 10.1088/1361-6528/ac1afc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Ruthenium(II) polypyridyl complexes (Ru) show high anti-tumor activity, but their poor solubility and low biocompatibility impede their use in anti-tumor therapy. Here,we circumvented the problem of low solubility by encapsulating the Ru in thermosensitive liposomes (LTSLs) and used gold nanorods (Au NRs) modified on the surface of the liposomes to permit the precise release of Ru at the tumor site. A facile and simple method was developed to synthesize Ru-loaded Au NR-decorated LTSL (Au@LTSL-Ru NPs). The loaded Au NRs improved the anti-tumor effect of Ru and enhanced the photothermal therapeutic properties of the nanosystem. A characterization experiment indicated that the average particle size of Au@LTSL-Ru was approximately 300 nm and that the Au NRs were successfully modified on the surface of LTSL. In thein vitroanti-tumor test, Au@LTSL-Ru and NIR significantly inhibited the proliferation of SGC-7901 cells. The IC50value of Au@LTSL-Ru + NIR was 7.1 ± 1.2μM (13μg ml-1), and the inhibition rate was greater than 90% when the concentration reached 30μg ml-1.In vivostudies revealed that Au@LTSL-Ru and NIR had a significant inhibitory effect on subcutaneous tumor tissues derived from SGC-7901 cells. Analysis of histopathology and immunocytotoxicity indicated that Au@LTSL-Ru has fewer side effects and high biocompatibility. Our results confirm that Au@LTSL-Ru can effectively inhibit tumor growth and aid the development of Ru for use in the thermal response in anti-tumor activity research.
Collapse
Affiliation(s)
- Longbao Zhu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Zhao Kuang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Wanzhen Li
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, Peoples Republic of China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, People's Republic of China
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| |
Collapse
|
25
|
Jahnke K, Ritzmann N, Fichtler J, Nitschke A, Dreher Y, Abele T, Hofhaus G, Platzman I, Schröder RR, Müller DJ, Spatz JP, Göpfrich K. Proton gradients from light-harvesting E. coli control DNA assemblies for synthetic cells. Nat Commun 2021; 12:3967. [PMID: 34172734 PMCID: PMC8233306 DOI: 10.1038/s41467-021-24103-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Bottom-up and top-down approaches to synthetic biology each employ distinct methodologies with the common aim to harness living systems. Here, we realize a strategic merger of both approaches to convert light into proton gradients for the actuation of synthetic cellular systems. We genetically engineer E. coli to overexpress the light-driven inward-directed proton pump xenorhodopsin and encapsulate them in artificial cell-sized compartments. Exposing the compartments to light-dark cycles, we reversibly switch the pH by almost one pH unit and employ these pH gradients to trigger the attachment of DNA structures to the compartment periphery. For this purpose, a DNA triplex motif serves as a nanomechanical switch responding to the pH-trigger of the E. coli. When DNA origami plates are modified with the pH-sensitive triplex motif, the proton-pumping E. coli can trigger their attachment to giant unilamellar lipid vesicles (GUVs) upon illumination. A DNA cortex is formed upon DNA origami polymerization, which sculpts and deforms the GUVs. We foresee that the combination of bottom-up and top down approaches is an efficient way to engineer synthetic cells.
Collapse
Affiliation(s)
- Kevin Jahnke
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Noah Ritzmann
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Julius Fichtler
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Anna Nitschke
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Yannik Dreher
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Tobias Abele
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Götz Hofhaus
- Centre for Advanced Materials, Heidelberg, Germany
| | - Ilia Platzman
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Heidelberg, Germany
| | | | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - Joachim P Spatz
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, Heidelberg, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
| | - Kerstin Göpfrich
- Biophysical Engineering Group, Max Planck Institute for Medical Research, Heidelberg, Germany.
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
26
|
Zhang T, Su M, Liu M, Tao M, Yang Y, Liu C, Zeng X, Pan D, Wu Z, Guo Y. Optimization of Encapsulation Using Milk Polar Lipid Liposomes with S-Layer Protein and Transport Study of the ACE-Inhibitory Peptide RLSFNP. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7049-7056. [PMID: 34132090 DOI: 10.1021/acs.jafc.1c02216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The purpose of this study is to develop a new type of nanodrug delivery material by modifying milk polar lipid (MPL) liposomes with the S-layer protein. LIP-RLSFNP (MPL liposomes encapsulating RLSFNP (Arg-Leu-Ser-Phe-Asn-Pro)) and SLP-LIP-RLSFNP (S-layer protein-modified LIP-RLSFNP) were prepared and characterized by transmission electron microscopy, Fourier transform infrared spectroscopy, confocal laser scanning microscopy, surface plasmon resonance, and mastersizer dynamic light scattering measurements. The results showed that the S-layer protein could modify the surface of MPL liposomes, stabilize the shape of the vesicles, and improve the resistance to external interference. Furthermore, SLP-LIP-RLSFNP showed better performance in in vitro and in vivo experiments compared with LIP-RLSFNP in terms of promoting absorption and delayed release. The findings suggested that MPL liposomes modified with the S-layer protein have potential for use as an effective delivery system for therapeutic proteins and peptides.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mi Su
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mingzhen Liu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Mingxuan Tao
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Yao Yang
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Chen Liu
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| | - Xiaoqun Zeng
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Daodong Pan
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Zhen Wu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Yuxing Guo
- Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210097, Jiangsu, P. R. China
| |
Collapse
|
27
|
Long K, Liu Y, Li Y, Wang W. Self-assembly of trigonal building blocks into nanostructures: molecular design and biomedical applications. J Mater Chem B 2021; 8:6739-6752. [PMID: 32686806 DOI: 10.1039/d0tb01128b] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Trigonal molecules have a special triskelion structure similar to clathrin protein, providing great inspiration for constructing artificial nanoassemblies. To date, various synthetic trigonal conjugates have been designed for supramolecular self-assembly, which have demonstrated versatile and controllable self-assembly ability in materials science. Here we will review the design of trigonal (sometimes called three-legged, tripodal, C3-symmetric, or triskelion) building blocks that can self-assemble into various nanostructures and discuss the biomedical applications of the self-assembled nanomaterials.
Collapse
Affiliation(s)
- Kaiqi Long
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | | | | | | |
Collapse
|
28
|
De Leo V, Milano F, Agostiano A, Catucci L. Recent Advancements in Polymer/Liposome Assembly for Drug Delivery: From Surface Modifications to Hybrid Vesicles. Polymers (Basel) 2021; 13:1027. [PMID: 33810273 PMCID: PMC8037206 DOI: 10.3390/polym13071027] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Liposomes are consolidated and attractive biomimetic nanocarriers widely used in the field of drug delivery. The structural versatility of liposomes has been exploited for the development of various carriers for the topical or systemic delivery of drugs and bioactive molecules, with the possibility of increasing their bioavailability and stability, and modulating and directing their release, while limiting the side effects at the same time. Nevertheless, first-generation vesicles suffer from some limitations including physical instability, short in vivo circulation lifetime, reduced payload, uncontrolled release properties, and low targeting abilities. Therefore, liposome preparation technology soon took advantage of the possibility of improving vesicle performance using both natural and synthetic polymers. Polymers can easily be synthesized in a controlled manner over a wide range of molecular weights and in a low dispersity range. Their properties are widely tunable and therefore allow the low chemical versatility typical of lipids to be overcome. Moreover, depending on their structure, polymers can be used to create a simple covering on the liposome surface or to intercalate in the phospholipid bilayer to give rise to real hybrid structures. This review illustrates the main strategies implemented in the field of polymer/liposome assembly for drug delivery, with a look at the most recent publications without neglecting basic concepts for a simple and complete understanding by the reader.
Collapse
Affiliation(s)
- Vincenzo De Leo
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Francesco Milano
- Istituto di Scienze delle Produzioni Alimentari (ISPA), Consiglio Nazionale delle Ricerche (CNR), S.P. Lecce-Monteroni, Ecotekne, 73100 Lecce, Italy;
| | - Angela Agostiano
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Lucia Catucci
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| |
Collapse
|
29
|
Czekalska MA, Jacobs AMJ, Toprakcioglu Z, Kong L, Baumann KN, Gang H, Zubaite G, Ye R, Mu B, Levin A, Huck WTS, Knowles TPJ. One-Step Generation of Multisomes from Lipid-Stabilized Double Emulsions. ACS APPLIED MATERIALS & INTERFACES 2021; 13:6739-6747. [PMID: 33522221 DOI: 10.1021/acsami.0c16019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Multisomes are multicompartmental structures formed by a lipid-stabilized network of aqueous droplets, which are contained by an outer oil phase. These biomimetic structures are emerging as a versatile platform for soft matter and synthetic biology applications. While several methods for producing multisomes have been described, including microfluidic techniques, approaches for generating biocompatible, monodisperse multisomes in a reproducible manner remain challenging to implement due to low throughput and complex device fabrication. Here, we report on a robust method for the dynamically controlled generation of multisomes with controllable sizes and high monodispersity from lipid-based double emulsions. The described microfluidic approach entails the use of three different phases forming a water/oil/water (W/O/W) double emulsion stabilized by lipid layers. We employ a gradient of glycerol concentration between the inner core and outer phase to drive the directed osmosis, allowing the swelling of lamellar lipid layers resulting in the formation of small aqueous daughter droplets at the interface of the inner aqueous core. By adding increasing concentrations of glycerol to the outer aqueous phase and subsequently varying the osmotic gradient, we show that key structural parameters, including the size of the internal droplets, can be specifically controlled. Finally, we show that this approach can be used to generate multisomes encapsulating small-molecule cargo, with potential applications in synthetic biology, drug delivery, and as carriers for active materials in the food and cosmetics industries.
Collapse
Affiliation(s)
- Magdalena A Czekalska
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, Warsaw 01-224, Poland
| | - Anne M J Jacobs
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Zenon Toprakcioglu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Lingling Kong
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- State Key Laboratory of Bioreactor Engineering and Applied Chemistry Institute, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Kevin N Baumann
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Hongze Gang
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- State Key Laboratory of Bioreactor Engineering and Applied Chemistry Institute, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Greta Zubaite
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Ruqiang Ye
- State Key Laboratory of Bioreactor Engineering and Applied Chemistry Institute, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Bozhong Mu
- State Key Laboratory of Bioreactor Engineering and Applied Chemistry Institute, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology, Shanghai 200237, China
| | - Aviad Levin
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 AJ, The Netherlands
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, J. J. Thomson Avenue, CB2 0HE Cambridge, United Kingdom
| |
Collapse
|
30
|
Wu H, Chen H, Hsu CY, Yeh C, Hsu H, Cheng C. Discotic material hexakis(4‐carboxyphenylethynyl)benzene inhibits
Escherichia coli
growth via the glycolysis pathway. J CHIN CHEM SOC-TAIP 2021. [DOI: 10.1002/jccs.202000508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Hsiu‐Hui Wu
- Department of Applied Chemistry National Chia‐Yi University Chia‐Yi City Taiwan, ROC
| | - Ho‐Lun Chen
- Department of Applied Chemistry National Chia‐Yi University Chia‐Yi City Taiwan, ROC
| | - Chih Ying Hsu
- Department of Applied Chemistry National Chia‐Yi University Chia‐Yi City Taiwan, ROC
| | - Chih‐Ling Yeh
- Department of Applied Chemistry National Chia‐Yi University Chia‐Yi City Taiwan, ROC
| | - Hsiu‐Fu Hsu
- Department of Chemistry Tamkang University New Taipei City Taiwan, ROC
| | - Chien‐Chung Cheng
- Department of Applied Chemistry National Chia‐Yi University Chia‐Yi City Taiwan, ROC
| |
Collapse
|
31
|
Higashi SL, Hirosawa KM, Suzuki KGN, Matsuura K, Ikeda M. One-Pot Construction of Multicomponent Supramolecular Materials Comprising Self-Sorted Supramolecular Architectures of DNA and Semi-Artificial Glycopeptides. ACS APPLIED BIO MATERIALS 2020; 3:9082-9092. [DOI: 10.1021/acsabm.0c01316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sayuri L. Higashi
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Koichiro M. Hirosawa
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kenichi G. N. Suzuki
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Masato Ikeda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho,
Chikusa-ku, Nagoya 464-8603, Japan
| |
Collapse
|
32
|
Rajwar A, Morya V, Kharbanda S, Bhatia D. DNA Nanodevices to Probe and Program Membrane Organization, Dynamics, and Applications. J Membr Biol 2020; 253:577-587. [DOI: 10.1007/s00232-020-00154-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/07/2020] [Indexed: 12/18/2022]
|
33
|
Bhalla N, Pan Y, Yang Z, Payam AF. Opportunities and Challenges for Biosensors and Nanoscale Analytical Tools for Pandemics: COVID-19. ACS NANO 2020; 14:7783-7807. [PMID: 32551559 PMCID: PMC7319134 DOI: 10.1021/acsnano.0c04421] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/18/2020] [Indexed: 05/05/2023]
Abstract
Biosensors and nanoscale analytical tools have shown huge growth in literature in the past 20 years, with a large number of reports on the topic of 'ultrasensitive', 'cost-effective', and 'early detection' tools with a potential of 'mass-production' cited on the web of science. Yet none of these tools are commercially available in the market or practically viable for mass production and use in pandemic diseases such as coronavirus disease 2019 (COVID-19). In this context, we review the technological challenges and opportunities of current bio/chemical sensors and analytical tools by critically analyzing the bottlenecks which have hindered the implementation of advanced sensing technologies in pandemic diseases. We also describe in brief COVID-19 by comparing it with other pandemic strains such as that of severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) for the identification of features that enable biosensing. Moreover, we discuss visualization and characterization tools that can potentially be used not only for sensing applications but also to assist in speeding up the drug discovery and vaccine development process. Furthermore, we discuss the emerging monitoring mechanism, namely wastewater-based epidemiology, for early warning of the outbreak, focusing on sensors for rapid and on-site analysis of SARS-CoV2 in sewage. To conclude, we provide holistic insights into challenges associated with the quick translation of sensing technologies, policies, ethical issues, technology adoption, and an overall outlook of the role of the sensing technologies in pandemics.
Collapse
Affiliation(s)
- Nikhil Bhalla
- Nanotechnology
and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, Shore Road, BT37
0QB Jordanstown, Northern Ireland, United Kingdom
- Healthcare
Technology Hub, Ulster University, Shore Road, BT37 0QB Jordanstown, Northern
Ireland, United Kingdom
| | - Yuwei Pan
- Cranfield
Water Science Institute, Cranfield University, Cranfield, Bedfordshire MK43 0AL, United Kingdom
| | - Zhugen Yang
- Cranfield
Water Science Institute, Cranfield University, Cranfield, Bedfordshire MK43 0AL, United Kingdom
| | - Amir Farokh Payam
- Nanotechnology
and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, Shore Road, BT37
0QB Jordanstown, Northern Ireland, United Kingdom
- Healthcare
Technology Hub, Ulster University, Shore Road, BT37 0QB Jordanstown, Northern
Ireland, United Kingdom
| |
Collapse
|