1
|
Shmakov SV, Sosnovitskaia ZP, Makhneva EA, Anikina MA, Kuznetsov A, Kondratev VM, Solomonov N, Boitsov VM, Fedorov VV, Mukhin IS, Bukatin AS, Bolshakov AD. Cells have the ability to break and chemically modify GaP(As) nanowires. NANOSCALE 2024. [PMID: 39440788 DOI: 10.1039/d4nr02513j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Semiconductor nanowires are known for their unusual geometry, providing unique electronic and optical properties. Substrates with vertical nanowires have highly non-uniform surfaces, which are attractive in terms of the study of live cells that can interact and be labeled with the wires. Despite several previous works studying cells cultivated over nanowires, questions regarding cell rupture and interaction with the wires remain open. Here, we demonstrate that nanowires can not only penetrate the cell membrane, but even be broken by a cell and trapped inside it. Even after mechanical poration of the membrane manifested by the efficient transfection and delivery of a fluorescent protein encoding plasmid, the cells are found to be viable for 7 days of incubation. The endocytosed wires are then aligned along the nucleus periphery and ousted to pseudopodia with the formation of nanowire-rich fibrils as a result of complex intracellular processes. We demonstrate that endocytosis of the wires may lead to their chemical modification manifested by the red shift of the luminescence spectra. Analysis of the wires' breakdown reveals that the cells can generate forces as high as several hundreds of nN. Using this work, we demonstrate several phenomena with the potential to be used in intriguing methods for intracellular visualization and the development of biointerfaces.
Collapse
Affiliation(s)
- Stanislav V Shmakov
- Faculty of Physics, St Petersburg State University, Universitetskaya Emb. 7-9, 199034 St Petersburg, Russia.
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
| | | | | | - Maria A Anikina
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russia
| | - Alexey Kuznetsov
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russia
| | - Valeriy M Kondratev
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russia
| | - Nikita Solomonov
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
| | - Vitali M Boitsov
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
| | | | - Ivan S Mukhin
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Higher School of Engineering Physics, Peter the Great Saint Petersburg Polytechnic University, Politekhnicheskaya 29, Saint Petersburg 195251, Russia
| | - Anton S Bukatin
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Institute for Analytical Instrumentation of the RAS, St Petersburg, 190103, Russia
| | - Alexey D Bolshakov
- Faculty of Physics, St Petersburg State University, Universitetskaya Emb. 7-9, 199034 St Petersburg, Russia.
- Alferov University, Khlopina 8/3, St Petersburg, 194021, Russia.
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russia
- Laboratory of Advanced Functional Materials, Yerevan State University, Yerevan 0025, Armenia
| |
Collapse
|
2
|
Liu X, Jiang J, Liu J, Yang H, Huang Z, Deng C, Li Y, Shang L, Wang X, Xie X, Wang J. Nanoneedle Array-Electroporation Facilitates Intranuclear Ribonucleoprotein Delivery and High Throughput Gene Editing. Adv Healthc Mater 2024:e2400645. [PMID: 39240050 DOI: 10.1002/adhm.202400645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/24/2024] [Indexed: 09/07/2024]
Abstract
Dendritic cells (DCs) are critical regulators of T cell immunity, with immense therapeutic potential against tumors and autoimmune diseases. Efficient gene editing in DCs is crucial for understanding their regulatory mechanisms and maximizing their therapeutic efficacy. However, DCs are notoriously difficult to transfect, posing a major bottleneck for conventional DNA and RNA-based editing approaches. Microneedle-mediated injection of Cas9/sgRNA ribonucleoprotein (RNP) directly into the nucleus, akin to gene editing in reproductive cells, offers promise but suffers from limitations in scalability. Here, an intranuclear delivery system using a hollow nanoneedle array (HNA) combined with nano-electroporation is developed. The 2 µm-high HNA physically reaches the nucleus, positioning the nuclear envelope and plasma membrane in close proximity at the tip. Transient electronic pulses then induce simultaneous perforations across all 3 membranes, enabling direct RNP delivery into the nucleus. This HNA-based system achieves efficient knockout of genes like PD-L1 in primary DCs, demonstrating its potential as a powerful tool for gene editing in DCs and other hard-to-transfect cells.
Collapse
Affiliation(s)
- Xinmin Liu
- Center for Reproductive Medicine and Department of Gynecology & Obstetrics, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Juan Jiang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Jing Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510080, P. R. China
| | - Hao Yang
- School of Life Science, Liaoning University, Shenyang, 110036, P. R. China
| | - Zhangping Huang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Caiguanxi Deng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Yongyong Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Liru Shang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xiafeng Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xi Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510080, P. R. China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| |
Collapse
|
3
|
Yee Mon KJ, Kim S, Dai Z, West JD, Zhu H, Jain R, Grimson A, Rudd BD, Singh A. Functionalized nanowires for miRNA-mediated therapeutic programming of naïve T cells. NATURE NANOTECHNOLOGY 2024; 19:1190-1202. [PMID: 38684809 PMCID: PMC11330359 DOI: 10.1038/s41565-024-01649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/13/2024] [Indexed: 05/02/2024]
Abstract
Cellular programming of naïve T cells can improve the efficacy of adoptive T-cell therapy. However, the current ex vivo engineering of T cells requires the pre-activation of T cells, which causes them to lose their naïve state. In this study, cationic-polymer-functionalized nanowires were used to pre-program the fate of primary naïve CD8+ T cells to achieve a therapeutic response in vivo. This was done by delivering single or multiple microRNAs to primary naïve mouse and human CD8+ T cells without pre-activation. The use of nanowires further allowed for the delivery of large, whole lentiviral particles with potential for long-term integration. The combination of deletion and overexpression of miR-29 and miR-130 impacted the ex vivo T-cell differentiation fate from the naïve state. The programming of CD8+ T cells using nanowire-delivered co-delivery of microRNAs resulted in the modulation of T-cell fitness by altering the T-cell proliferation, phenotypic and transcriptional regulation, and secretion of effector molecules. Moreover, the in vivo adoptive transfer of murine CD8+ T cells programmed through the nanowire-mediated dual delivery of microRNAs provided enhanced immune protection against different types of intracellular pathogen (influenza and Listeria monocytogenes). In vivo analyses demonstrated that the simultaneous alteration of miR-29 and miR-130 levels in naïve CD8+ T cells reduces the persistence of canonical memory T cells whereas increases the population of short-lived effector T cells. Nanowires could potentially be used to modulate CD8+ T-cell differentiation and achieve a therapeutic response in vivo without the need for pre-activation.
Collapse
Affiliation(s)
- Kristel J Yee Mon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Sungwoong Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA
| | - Zhonghao Dai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jessica D West
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Hongya Zhu
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew Grimson
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA.
| | - Ankur Singh
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
4
|
Hu T, Kumar AR, Luo Y, Tay A. Automating CAR-T Transfection with Micro and Nano-Technologies. SMALL METHODS 2024; 8:e2301300. [PMID: 38054597 DOI: 10.1002/smtd.202301300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Cancer poses a significant health challenge, with traditional treatments like surgery, radiotherapy, and chemotherapy often lacking in cell specificity and long-term curative potential. Chimeric antigen receptor T cell (CAR-T) therapy,utilizing genetically engineered T cells to target cancer cells, is a promising alternative. However, its high cost limits widespread application. CAR-T manufacturing process encompasses three stages: cell isolation and activation, transfection, and expansion.While the first and last stages have straightforward, commercially available automation technologies, the transfection stage lags behind. Current automated transfection relies on viral vectors or bulk electroporation, which have drawbacks such as limited cargo capacity and significant cell disturbance. Conversely, micro and nano-tool methods offer higher throughput and cargo flexibility, yet their automation remains underexplored.In this perspective, the progress in micro and nano-engineering tools for CAR-T transfection followed by a discussion to automate them is described. It is anticipated that this work can inspire the community working on micro and nano transfection techniques to examine how their protocols can be automated to align with the growing interest in automating CAR-T manufacturing.
Collapse
Affiliation(s)
- Tianmu Hu
- Engineering Science Programme, National University of Singapore, Singapore, 117575, Singapore
| | - Arun Rk Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yikai Luo
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Tissue Engineering Programme, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
5
|
Wang C, Gu C, Popp C, Vashisth P, Mustfa SA, Martella DA, Spiteri C, McLennan S, Sun N, Riddle M, Eide CR, Parsons M, Tolar J, McGrath JA, Chiappini C. Integrating Porous Silicon Nanoneedles within Medical Devices for Nucleic Acid Nanoinjection. ACS NANO 2024; 18:14938-14953. [PMID: 38726598 PMCID: PMC11171749 DOI: 10.1021/acsnano.4c00206] [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: 01/05/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 06/12/2024]
Abstract
Porous silicon nanoneedles can interface with cells and tissues with minimal perturbation for high-throughput intracellular delivery and biosensing. Typically, nanoneedle devices are rigid, flat, and opaque, which limits their use for topical applications in the clinic. We have developed a robust, rapid, and precise substrate transfer approach to incorporate nanoneedles within diverse substrates of arbitrary composition, flexibility, curvature, transparency, and biodegradability. With this approach, we integrated nanoneedles on medically relevant elastomers, hydrogels, plastics, medical bandages, catheter tubes, and contact lenses. The integration retains the mechanical properties and transfection efficiency of the nanoneedles. Transparent devices enable the live monitoring of cell-nanoneedle interactions. Flexible devices interface with tissues for efficient, uniform, and sustained topical delivery of nucleic acids ex vivo and in vivo. The versatility of this approach highlights the opportunity to integrate nanoneedles within existing medical devices to develop advanced platforms for topical delivery and biosensing.
Collapse
Affiliation(s)
- Cong Wang
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Chenlei Gu
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Courtney Popp
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Priya Vashisth
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Salman Ahmad Mustfa
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Davide Alessandro Martella
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| | - Chantelle Spiteri
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Samuel McLennan
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Ningjia Sun
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
| | - Megan Riddle
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Cindy R. Eide
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Maddy Parsons
- Randall
Centre for Cell and Molecular Biophysics, King’s College London, SE1 1UL London, U.K.
| | - Jakub Tolar
- Department
of Pediatrics, Medical School, University
of Minnesota, Minneapolis, Minnesota 55455, United States
- Stem
Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - John A. McGrath
- St
John’s
Institute of Dermatology, King’s
College London, SE1 7EP London, U.K.
| | - Ciro Chiappini
- Centre
for Craniofacial and Regenerative Biology, King’s College London, SE1 9RT London, U.K.
- London
Centre for Nanotechnology, King’s
College London, WC2R 2LS London, U.K.
| |
Collapse
|
6
|
Zhao X, Mou C, Xu J, Cui W, Shi Y, Wang Y, Luo T, Guo W, Ye J, Chen W. Protection of Si Nanowires against A β Toxicity by the Inhibition of A β Aggregation. Molecules 2024; 29:1980. [PMID: 38731472 PMCID: PMC11085270 DOI: 10.3390/molecules29091980] [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: 03/26/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid beta (Aβ) plaques in the brain. Aβ1-42 is the main component of Aβ plaque, which is toxic to neuronal cells. Si nanowires (Si NWs) have the advantages of small particle size, high specific surface area, and good biocompatibility, and have potential application prospects in suppressing Aβ aggregation. In this study, we employed the vapor-liquid-solid (VLS) growth mechanism to grow Si NWs using Au nanoparticles as catalysts in a plasma-enhanced chemical vapor deposition (PECVD) system. Subsequently, these Si NWs were transferred to a phosphoric acid buffer solution (PBS). We found that Si NWs significantly reduced cell death in PC12 cells (rat adrenal pheochromocytoma cells) induced by Aβ1-42 oligomers via double staining with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and fluorescein diacetate/propyl iodide (FDA/PI). Most importantly, pre-incubated Si NWs largely prevented Aβ1-42 oligomer-induced PC12 cell death, suggesting that Si NWs exerts an anti-Aβ neuroprotective effect by inhibiting Aβ aggregation. The analysis of Fourier Transform Infrared (FTIR) results demonstrates that Si NWs reduce the toxicity of fibrils and oligomers by intervening in the formation of β-sheet structures, thereby protecting the viability of nerve cells. Our findings suggest that Si NWs may be a potential therapeutic agent for AD by protecting neuronal cells from the toxicity of Aβ1-42.
Collapse
Affiliation(s)
- Xuechun Zhao
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Chenye Mou
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Jiayi Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; (C.M.); (J.X.)
| | - Yijing Shi
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Yangzhe Wang
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| | - Tian Luo
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Wei Guo
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Jichun Ye
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China; (T.L.); (W.G.); (J.Y.)
| | - Wanghua Chen
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China; (X.Z.); (Y.S.); (Y.W.)
| |
Collapse
|
7
|
Xiang Y, Shi K, Li Y, Xue J, Tong Z, Li H, Li Z, Teng C, Fang J, Hu N. Active Micro-Nano-Collaborative Bioelectronic Device for Advanced Electrophysiological Recording. NANO-MICRO LETTERS 2024; 16:132. [PMID: 38411852 PMCID: PMC10899154 DOI: 10.1007/s40820-024-01336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 02/28/2024]
Abstract
The development of precise and sensitive electrophysiological recording platforms holds the utmost importance for research in the fields of cardiology and neuroscience. In recent years, active micro/nano-bioelectronic devices have undergone significant advancements, thereby facilitating the study of electrophysiology. The distinctive configuration and exceptional functionality of these active micro-nano-collaborative bioelectronic devices offer the potential for the recording of high-fidelity action potential signals on a large scale. In this paper, we review three-dimensional active nano-transistors and planar active micro-transistors in terms of their applications in electro-excitable cells, focusing on the evaluation of the effects of active micro/nano-bioelectronic devices on electrophysiological signals. Looking forward to the possibilities, challenges, and wide prospects of active micro-nano-devices, we expect to advance their progress to satisfy the demands of theoretical investigations and medical implementations within the domains of cardiology and neuroscience research.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China
| | - Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, People's Republic of China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People's Republic of China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China
| | - Zhicheng Tong
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Huiming Li
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, 523059, People's Republic of China.
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, 523059, People's Republic of China.
| | - Chong Teng
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322005, People's Republic of China.
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, People's Republic of China.
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
8
|
Kim H, Kim S, Lim H, Chung AJ. Expanding CAR-T cell immunotherapy horizons through microfluidics. LAB ON A CHIP 2024; 24:1088-1120. [PMID: 38174732 DOI: 10.1039/d3lc00622k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapies have revolutionized cancer treatment, particularly in hematological malignancies. However, their application to solid tumors is limited, and they face challenges in safety, scalability, and cost. To enhance current CAR-T cell therapies, the integration of microfluidic technologies, harnessing their inherent advantages, such as reduced sample consumption, simplicity in operation, cost-effectiveness, automation, and high scalability, has emerged as a powerful solution. This review provides a comprehensive overview of the step-by-step manufacturing process of CAR-T cells, identifies existing difficulties at each production stage, and discusses the successful implementation of microfluidics and related technologies in addressing these challenges. Furthermore, this review investigates the potential of microfluidics-based methodologies in advancing cell-based therapy across various applications, including solid tumors, next-generation CAR constructs, T-cell receptors, and the development of allogeneic "off-the-shelf" CAR products.
Collapse
Affiliation(s)
- Hyelee Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Suyeon Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Hyunjung Lim
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Aram J Chung
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
- School of Biomedical Engineering, Korea University, 02841 Seoul, Republic of Korea.
- MxT Biotech, 04785 Seoul, Republic of Korea
| |
Collapse
|
9
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
10
|
Gao Q, Liu J, Wang M, Liu X, Jiang Y, Su J. Biomaterials regulates BMSCs differentiation via mechanical microenvironment. BIOMATERIALS ADVANCES 2024; 157:213738. [PMID: 38154401 DOI: 10.1016/j.bioadv.2023.213738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Bone mesenchymal stem cells (BMSCs) are crucial for bone tissue regeneration, the mechanical microenvironment of hard tissues, including bone and teeth, significantly affects the osteogenic differentiation of BMSCs. Biomaterials may mimic the microenvironment of the extracellular matrix and provide mechanical signals to regulate BMSCs differentiation via inducing the secretion of various intracellular factors. Biomaterials direct the differentiation of BMSCs via mechanical signals, including tension, compression, shear, hydrostatic pressure, stiffness, elasticity, and viscoelasticity, which can be transmitted to cells through mechanical signalling pathways. Besides, biomaterials with piezoelectric effects regulate BMSCs differentiation via indirect mechanical signals, such as, electronic signals, which are transformed from mechanical stimuli by piezoelectric biomaterials. Mechanical stimulation facilitates achieving vectored stem cell fate regulation, while understanding the underlying mechanisms remains challenging. Herein, this review summarizes the intracellular factors, including translation factors, epigenetic modifications, and miRNA level, as well as the extracellular factor, including direct and indirect mechanical signals, which regulate the osteogenic differentiation of BMSCs. Besides, this review will also give a comprehensive summary about how mechanical stimuli regulate cellular behaviours, as well as how biomaterials promote the osteogenic differentiation of BMSCs via mechanical microenvironments. The cellular behaviours and activated signal pathways will give more implications for the design of biomaterials with superior properties for bone tissue engineering. Moreover, it will also provide inspiration for the construction of bone organoids which is a useful tool for mimicking in vivo bone tissue microenvironments.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Jinlong Liu
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China
| | - Xiangfei Liu
- Department of Orthopedics, Shanghai Zhongye Hospital, NO. 456 Chunlei Road, Shanghai 200941, PR China.
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; Organoid Research Centre, Shanghai University, NO.333 Nanchen Road, Shanghai 200444, PR China; National Centre for Translational Medicine (Shanghai) SHU Branch, NO.333 Nanchen Road, Shanghai University, Shanghai 200444, PR China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, NO.1665 Kongjiang Road, Shanghai 200092, PR China.
| |
Collapse
|
11
|
Cournut A, Moustiez P, Coffinier Y, Enjalbal C, Bich C. Innovative SALDI mass spectrometry analysis for Alzheimer's disease synthetic peptides detection. Talanta 2024; 268:125357. [PMID: 37951181 DOI: 10.1016/j.talanta.2023.125357] [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: 07/04/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/13/2023]
Abstract
Alzheimer's disease (AD) is nowadays the prominent cause of senile dementia. This pathology is characterized by aggregation of neurofibrillary tangles in cells and by the accumulation of amyloid plaques in the brain. Noteworthy, a phosphorylated protein (tau protein) and a peptide presenting two overlapping sequences of 40 or 42 residues named β-amyloid peptides 1-40 (Aβ 1-40) and 1-42 (Aβ 1-42), respectively, were related to such deleterious phenomena. Singularly, the neurotoxicity was primarily attributed to the amyloid peptide Aβ 1-42 form due to its capacity to fold into beta-sheets rendering it insoluble thus causing subsequent aggregation and accumulation in vivo. Regarding AD diagnosis relying on mass spectrometry, Aβ 1-42 and/or Aβ 1-40 were considered as relevant biomarkers being measured in cerebrospinal fluids (CSF), blood and urine. Under that context, we aimed at implementing an innovative method to evidence the depletion of circulating Aβ 1-42 amyloid peptide compared to the shorter Aβ 1-40 form indicating a pathologic state. We investigated Surface-Assisted Laser Desorption/Ionization Mass Spectrometry (SALDI-MS) in order to monitor the Aβ 1-42/Aβ 1-40 ratio without any prior sample treatment or enrichment. Taking into account that β-amyloid peptide and 1-42 can aggregate into beta-sheets depending on the experimental conditions, specific attention was devoted to sample integrity monitoring performed by circular dichroism experiments during SALDI-MS method development.
Collapse
Affiliation(s)
- Aline Cournut
- Univ Montpellier, IBMM, CNRS, ENSCM, Montpellier, France
| | - Paul Moustiez
- Univ Lille, IEMN, UMR CNRS 8520, Villeneuve d'Ascq, France
| | | | | | - Claudia Bich
- Univ Montpellier, IBMM, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
12
|
Zhao P, Song S, He Z, Dai G, Liu D, Shen J, Asakawa T, Zheng M, Lu H. Development of a novel cholesterol tag-based system for trans-membrane transport of protein drugs. Biosci Trends 2024; 17:503-507. [PMID: 38072446 DOI: 10.5582/bst.2023.01285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
The main technological difficulties of developing an intracellular (transmembrane) transport system for protein drugs lie in two points: i) overcoming the barriers in the cellular membrane, and ii) loading enough protein drugs, and particularly high-dose proteins, into particles. To address these two technological problems, we recently developed a novel cholesterol tag (C-Tag)-based transmembrane transport system. This pilot study found that the C-Tag dramatically improved the cellular uptake of Fab (902-fold, vs. Fab alone) into living cells, indicating that it successfully achieved transmembrane transport. Moreover, C-Tag-mediated membrane transport was verified using micron-scale large unilamellar vesicles (LUVs, approximately 1.5 μm)-based particles. The C-Tagged Fab was able to permeate the liposomal bilayer and it greatly enhanced (a 10.1-fold increase vs. Fab alone) internalization of proteins into the LUV-based particles, indicating that the C-Tag loaded enough proteins into particles for use of high-dose proteins. Accordingly, we established a novel C-Tag-based transport system that has overcome the known technological difficulties of protein transmembrane delivery, and this might be a useful technology for drug development in the future.
Collapse
Affiliation(s)
- Pengfei Zhao
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, China
| | - Shuo Song
- Shenzhen Samii Medical Center, Shenzhen, Guangdong, China
| | - Zhuojun He
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Guiqin Dai
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Deliang Liu
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Jiayin Shen
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Mingbin Zheng
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Shenzhen Samii Medical Center, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Park R, Kang MS, Heo G, Shin YC, Han DW, Hong SW. Regulated Behavior in Living Cells with Highly Aligned Configurations on Nanowrinkled Graphene Oxide Substrates: Deep Learning Based on Interplay of Cellular Contact Guidance. ACS NANO 2024; 18:1325-1344. [PMID: 38099607 DOI: 10.1021/acsnano.2c09815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Micro-/nanotopographical cues have emerged as a practical and promising strategy for controlling cell fate and reprogramming, which play a key role as biophysical regulators in diverse cellular processes and behaviors. Extracellular biophysical factors can trigger intracellular physiological signaling via mechanotransduction and promote cellular responses such as cell adhesion, migration, proliferation, gene/protein expression, and differentiation. Here, we engineered a highly ordered nanowrinkled graphene oxide (GO) surface via the mechanical deformation of an ultrathin GO film on an elastomeric substrate to observe specific cellular responses based on surface-mediated topographical cues. The ultrathin GO film on the uniaxially prestrained elastomeric substrate through self-assembly and subsequent compressive force produced GO nanowrinkles with periodic amplitude. To examine the acute cellular behaviors on the GO-based cell interface with nanostructured arrays of wrinkles, we cultured L929 fibroblasts and HT22 hippocampal neuronal cells. As a result, our developed cell-culture substrate obviously provided a directional guidance effect. In addition, based on the observed results, we adapted a deep learning (DL)-based data processing technique to precisely interpret the cell behaviors on the nanowrinkled GO surfaces. According to the learning/transfer learning protocol of the DL network, we detected cell boundaries, elongation, and orientation and quantitatively evaluated cell velocity, traveling distance, displacement, and orientation. The presented experimental results have intriguing implications such that the nanotopographical microenvironment could engineer the living cells' morphological polarization to assemble them into useful tissue chips consisting of multiple cell types.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Gyeonghwa Heo
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio 44195, United States
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
14
|
Lyu M, Yazdi M, Lin Y, Höhn M, Lächelt U, Wagner E. Receptor-Targeted Dual pH-Triggered Intracellular Protein Transfer. ACS Biomater Sci Eng 2024; 10:99-114. [PMID: 35802884 DOI: 10.1021/acsbiomaterials.2c00476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein therapeutics are of widespread interest due to their successful performance in the current pharmaceutical and medical fields, even though their broad applications have been hindered by the lack of an efficient intracellular delivery approach. Herein, we fabricated an active-targeted dual pH-responsive delivery system with favorable tumor cell entry augmented by extracellular pH-triggered charge reversal and tumor receptor targeting and pH-controlled endosomal release in a traceless fashion. As a traceable model protein, the enhanced green fluorescent protein (eGFP) bearing a nuclear localization signal was covalently coupled with a pH-labile traceless azidomethyl-methylmaleic anhydride (AzMMMan) linker followed by functionalization with different molar equivalents of two dibenzocyclooctyne-octa-arginine-cysteine (DBCO-R8C)-modified moieties: polyethylene glycol (PEG)-GE11 peptide for epidermal growth factor receptor-mediated targeting and melittin for endosomal escape. The cationic melittin domain was masked with tetrahydrophthalic anhydride revertible at mild acidic pH 6.5. At the optimally balanced ratio of functional units, the on-demand charge conversion at tumoral extracellular pH 6.5 in combination with GE11-mediated targeting triggered enhanced electrostatic cellular attraction by the R8C cell-penetrating peptides and melittin, as demonstrated by strongly enhanced cellular uptake. Successful endosomal release followed by nuclear localization of the eGFP cargo was obtained by taking advantage of melittin-mediated endosomal escape and rapid traceless release from the AzMMMan linker. The effectiveness of this multifunctional bioresponsive system suggests a promising strategy for delivery of protein drugs toward intracellular targets. A possible therapeutic relevance was indicated by an example of cytosolic delivery of cytochrome c initiating the apoptosis pathway to kill cancer cells.
Collapse
Affiliation(s)
- Meng Lyu
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
15
|
Wang H, Xue J, Li Y, Shi K, Fang J, Zheng J, Lyu X, Gao Z, Xu D, Hu N. Optimizing the Cell-Nanostructure Interface: Nanoconcave/Nanoconvex Device for Intracellular Recording of Cardiomyocytes. NANO LETTERS 2023; 23:11884-11891. [PMID: 38064276 DOI: 10.1021/acs.nanolett.3c03945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Nanostructures are powerful components for the development of high-performance nanodevices. Revealing and understanding the cell-nanostructure interface are essential for improving and guiding nanodevice design for investigations of cell physiology. For intracellular electrophysiological detection, the cell-nanostructure interface significantly affects the quality of recorded intracellular action potentials and the application of nanodevices in cardiology research and pharmacological screening. Most of the current investigations of biointerfaces focus on nanovertical structures, and few involve nanoconcave structures. Here, we design both nanoconvex and nanoconcave devices to perform intracellular electrophysiological recordings. The amplitude, signal-to-noise ratio, duration, and repeatability of the recorded intracellular electrophysiological signals provide a multifaceted characterization of the cell-nanostructure interface. We demonstrate that devices based on both convex and concave nanostructures can create tight coupling, which facilitates high-quality and stable intracellular recordings and paves the way for precise electrophysiological study.
Collapse
Affiliation(s)
- Hao Wang
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Xue
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Ying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Keda Shi
- Department of Lung Transplantation and General Thoracic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiaru Fang
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Jilin Zheng
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Xuelian Lyu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
| | - Zhigang Gao
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Dongxin Xu
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 310058, China
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| |
Collapse
|
16
|
Park W, Kim EM, Jeon Y, Lee J, Yi J, Jeong J, Kim B, Jeong BG, Kim DR, Kong H, Lee CH. Transparent Intracellular Sensing Platform with Si Needles for Simultaneous Live Imaging. ACS NANO 2023; 17:25014-25026. [PMID: 38059775 DOI: 10.1021/acsnano.3c07527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Vertically ordered Si needles are of particular interest for long-term intracellular recording owing to their capacity to infiltrate living cells with negligible damage and minimal toxicity. Such intracellular recordings could greatly benefit from simultaneous live cell imaging without disrupting their culture, contributing to an in-depth understanding of cellular function and activity. However, the use of standard live imaging techniques, such as inverted and confocal microscopy, is currently impeded by the opacity of Si wafers, typically employed for fabricating vertical Si needles. Here, we introduce a transparent intracellular sensing platform that combines vertical Si needles with a percolated network of Au-Ag nanowires on a transparent elastomeric substrate. This sensing platform meets all prerequisites for simultaneous intracellular recording and imaging, including electrochemical impedance, optical transparency, mechanical compliance, and cell viability. Proof-of-concept demonstrations of this sensing platform include monitoring electrical potentials in cardiomyocyte cells and in three-dimensionally engineered cardiovascular tissue, all while conducting live imaging with inverted and confocal microscopes. This sensing platform holds wide-ranging potential applications for intracellular research across various disciplines such as neuroscience, cardiology, muscle physiology, and drug screening.
Collapse
Affiliation(s)
- Woohyun Park
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Eun Mi Kim
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yale Jeon
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Junsang Lee
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jonghun Yi
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinheon Jeong
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bongjoong Kim
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Mechanical and System Design Engineering, Hongik University, Seoul 04066, Republic of Korea
| | - Byeong Guk Jeong
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dong Rip Kim
- School of Mechanical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chi Hwan Lee
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Materials Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
17
|
Guo X, Ma Y, Wang H, Yin H, Shi X, Chen Y, Gao G, Sun L, Wang J, Wang Y, Fan D. Status and developmental trends in recombinant collagen preparation technology. Regen Biomater 2023; 11:rbad106. [PMID: 38173768 PMCID: PMC10761200 DOI: 10.1093/rb/rbad106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Recombinant collagen is a pivotal topic in foundational biological research and epitomizes the application of critical bioengineering technologies. These technological advancements have profound implications across diverse areas such as regenerative medicine, organ replacement, tissue engineering, cosmetics and more. Thus, recombinant collagen and its preparation methodologies rooted in genetically engineered cells mark pivotal milestones in medical product research. This article provides a comprehensive overview of the current genetic engineering technologies and methods used in the production of recombinant collagen, as well as the conventional production process and quality control detection methods for this material. Furthermore, the discussion extends to foresee the strides in physical transfection and magnetic control sorting studies, envisioning an enhanced preparation of recombinant collagen-seeded cells to further fuel recombinant collagen production.
Collapse
Affiliation(s)
- Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Yuan Ma
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Hang Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Yiqin Chen
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Guobiao Gao
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Lei Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing 100081, China
| | - Jiadao Wang
- State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials & College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Daidi Fan
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710127, China
| |
Collapse
|
18
|
Shokouhi AR, Chen Y, Yoh HZ, Brenker J, Alan T, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Engineering Efficient CAR-T Cells via Electroactive Nanoinjection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304122. [PMID: 37434421 DOI: 10.1002/adma.202304122] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Jason Brenker
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Waurn Ponds, VIC, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia
| |
Collapse
|
19
|
Rahamathulla M, Murugesan S, Gowda DV, Alamri AH, Ahmed MM, Osmani RAM, Ramamoorthy S, Veeranna B. The Use of Nanoneedles in Drug Delivery: an Overview of Recent Trends and Applications. AAPS PharmSciTech 2023; 24:216. [PMID: 37857918 DOI: 10.1208/s12249-023-02661-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/15/2023] [Indexed: 10/21/2023] Open
Abstract
Nanoneedles (NN) are growing rapidly as a means of navigating biological membranes and delivering therapeutics intracellularly. Nanoneedle arrays (NNA) are among the most potential resources to achieve therapeutic effects by administration of drugs through the skin. Although this is based on well-established approaches, its implementations are rapidly developing as an important pharmaceutical and biological research phenomenon. This study intends to provide a broad overview of current NNA research, with an emphasis on existing approaches, applications, and types of compounds released by these systems. A nanoneedle-based delivery device with great spatial and temporal accuracy, minimal interference, and low toxicity could transfer biomolecules into living organisms. Due to its vast potential, NN has been widely used as a capable transportation system of many therapeutic active substances, from cancer therapy, vaccine delivery, cosmetics, and bio-sensing nanocarrier drugs to genes. The use of nanoneedles for drug delivery offers new opportunities for the rapid, targeted, and exact administration of biomolecules into cell membranes for high-resolution research of biological systems, and it can treat a wide range of biological challenges. As a result, the literature has analyzed existing patents to emphasize the status of NNA in biological applications.
Collapse
Affiliation(s)
- Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Santhosh Murugesan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, 570015, Karnataka, India
| | - D V Gowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, 570015, Karnataka, India
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, 570015, Karnataka, India.
| | - Sathish Ramamoorthy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, 570015, Karnataka, India
| | - Balamuralidhara Veeranna
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, 570015, Karnataka, India.
| |
Collapse
|
20
|
Bigham A, Raucci MG, Zheng K, Boccaccini AR, Ambrosio L. Oxygen-Deficient Bioceramics: Combination of Diagnosis, Therapy, and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302858. [PMID: 37259776 DOI: 10.1002/adma.202302858] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/15/2023] [Indexed: 06/02/2023]
Abstract
The journey of ceramics in medicine has been synchronized with an evolution from the first generation-alumina, zirconia, etc.-to the third -3D scaffolds. There is an up-and-coming member called oxygen-deficient or colored bioceramics, which have recently found their way through biomedical applications. The oxygen vacancy steers the light absorption toward visible and near infrared regions, making the colored bioceramics multifunctional-therapeutic, diagnostic, and regenerative. Oxygen-deficient bioceramics are capable of turning light into heat and reactive oxygen species for photothermal and photodynamic therapies, respectively, and concomitantly yield infrared and photoacoustic images. Different types of oxygen-deficient bioceramics have been recently developed through various synthesis routes. Some of them like TiO2- x , MoO3- x , and WOx have been more investigated for biomedical applications, whereas the rest have yet to be scrutinized. The most prominent advantage of these bioceramics over the other biomaterials is their multifunctionality endowed with a change in the microstructure. There are some challenges ahead of this category discussed at the end of the present review. By shedding light on this recently born bioceramics subcategory, it is believed that the field will undergo a big step further as these platforms are naturally multifunctional.
Collapse
Affiliation(s)
- Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials-National Research Council (IPCB-CNR), Viale J. F. Kennedy 54-Mostra d'Oltremare pad. 20, Naples, 80125, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, Naples, 80125, Italy
| | - Maria Grazia Raucci
- Institute of Polymers, Composites and Biomaterials-National Research Council (IPCB-CNR), Viale J. F. Kennedy 54-Mostra d'Oltremare pad. 20, Naples, 80125, Italy
| | - Kai Zheng
- Jiangsu Key Laboratory of Oral Diseases and Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Aldo R Boccaccini
- Institute for Biomaterials, University of Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials-National Research Council (IPCB-CNR), Viale J. F. Kennedy 54-Mostra d'Oltremare pad. 20, Naples, 80125, Italy
| |
Collapse
|
21
|
Xu H, Wu L, Xue Y, Yang T, Xiong T, Wang C, He S, Sun H, Cao Z, Liu J, Wang S, Li Z, Naeem A, Yin X, Zhang J. Advances in Structure Pharmaceutics from Discovery to Evaluation and Design. Mol Pharm 2023; 20:4404-4429. [PMID: 37552597 DOI: 10.1021/acs.molpharmaceut.3c00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Drug delivery systems (DDSs) play an important role in delivering active pharmaceutical ingredients (APIs) to targeted sites with a predesigned release pattern. The chemical and biological properties of APIs and excipients have been extensively studied for their contribution to DDS quality and effectiveness; however, the structural characteristics of DDSs have not been adequately explored. Structure pharmaceutics involves the study of the structure of DDSs, especially the three-dimensional (3D) structures, and its interaction with the physiological and pathological structure of organisms, possibly influencing their release kinetics and targeting abilities. A systematic overview of the structures of a variety of dosage forms, such as tablets, granules, pellets, microspheres, powders, and nanoparticles, is presented. Moreover, the influence of structures on the release and targeting capability of DDSs has also been discussed, especially the in vitro and in vivo release correlation and the structure-based organ- and tumor-targeting capabilities of particles with different structures. Additionally, an in-depth discussion is provided regarding the application of structural strategies in the DDSs design and evaluation. Furthermore, some of the most frequently used characterization techniques in structure pharmaceutics are briefly described along with their potential future applications.
Collapse
Affiliation(s)
- Huipeng Xu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Wu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Ministry of Education, Yantai University, Yantai 264005, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yanling Xue
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Ting Yang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ting Xiong
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Caifen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyu He
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyu Sun
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zeying Cao
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Liu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siwen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhe Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Abid Naeem
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xianzhen Yin
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 201602, China
| | - Jiwen Zhang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No.2 Tiantan Xili, Beijing 100050, China
| |
Collapse
|
22
|
Shokouhi AR, Chen Y, Yoh HZ, Murayama T, Suu K, Morikawa Y, Brenker J, Alan T, Voelcker NH, Elnathan R. Electroactive nanoinjection platform for intracellular delivery and gene silencing. J Nanobiotechnology 2023; 21:273. [PMID: 37592297 PMCID: PMC10433684 DOI: 10.1186/s12951-023-02056-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Jason Brenker
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Melbourne, VIC, 3216, Australia.
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Melbourne, VIC, 3216, Australia.
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia.
| |
Collapse
|
23
|
Suryadevara V, Hajipour MJ, Adams LC, Aissaoui NM, Rashidi A, Kiru L, Theruvath AJ, Huang C, Maruyama M, Tsubosaka M, Lyons JK, Wu W(E, Roudi R, Goodman SB, Daldrup‐Link HE. MegaPro, a clinically translatable nanoparticle for in vivo tracking of stem cell implants in pig cartilage defects. Theranostics 2023; 13:2710-2720. [PMID: 37215574 PMCID: PMC10196837 DOI: 10.7150/thno.82620] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
Rationale: Efficient labeling methods for mesenchymal stem cells (MSCs) are crucial for tracking and understanding their behavior in regenerative medicine applications, particularly in cartilage defects. MegaPro nanoparticles have emerged as a potential alternative to ferumoxytol nanoparticles for this purpose. Methods: In this study, we employed mechanoporation to develop an efficient labeling method for MSCs using MegaPro nanoparticles and compared their effectiveness with ferumoxytol nanoparticles in tracking MSCs and chondrogenic pellets. Pig MSCs were labeled with both nanoparticles using a custom-made microfluidic device, and their characteristics were analyzed using various imaging and spectroscopy techniques. The viability and differentiation capacity of labeled MSCs were also assessed. Labeled MSCs and chondrogenic pellets were implanted into pig knee joints and monitored using MRI and histological analysis. Results: MegaPro-labeled MSCs demonstrated shorter T2 relaxation times, higher iron content, and greater nanoparticle uptake compared to ferumoxytol-labeled MSCs, without significantly affecting their viability and differentiation capacity. Post-implantation, MegaPro-labeled MSCs and chondrogenic pellets displayed a strong hypointense signal on MRI with considerably shorter T2* relaxation times compared to adjacent cartilage. The hypointense signal of both MegaPro- and ferumoxytol-labeled chondrogenic pellets decreased over time. Histological evaluations showed regenerated defect areas and proteoglycan formation with no significant differences between the labeled groups. Conclusion: Our study demonstrates that mechanoporation with MegaPro nanoparticles enables efficient MSC labeling without affecting viability or differentiation. MegaPro-labeled cells show enhanced MRI tracking compared to ferumoxytol-labeled cells, emphasizing their potential in clinical stem cell therapies for cartilage defects.
Collapse
Affiliation(s)
- Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Mohammad Javad Hajipour
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa C. Adams
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Nour Mary Aissaoui
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Ali Rashidi
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Ashok J. Theruvath
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Ching‐Hsin Huang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Masahiro Maruyama
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masanori Tsubosaka
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer K. Lyons
- Department of Veterinary Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wei (Emma) Wu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B. Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Heike E. Daldrup‐Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
24
|
El-Yadri M, El Hamdaoui J, Aghoutane N, Pérez LM, Baskoutas S, Laroze D, Díaz P, Feddi EM. Optoelectronic Properties of a Cylindrical Core/Shell Nanowire: Effect of Quantum Confinement and Magnetic Field. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1334. [PMID: 37110919 PMCID: PMC10141194 DOI: 10.3390/nano13081334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 06/19/2023]
Abstract
This study investigates the effect of quantum size and an external magnetic field on the optoelectronic properties of a cylindrical AlxGa1-xAs/GaAs-based core/shell nanowire. We used the one-band effective mass model to describe the Hamiltonian of an interacting electron-donor impurity system and employed two numerical methods to calculate the ground state energies: the variational and finite element methods. With the finite confinement barrier at the interface between the core and the shell, the cylindrical symmetry of the system revealed proper transcendental equations, leading to the concept of the threshold core radius. Our results show that the optoelectronic properties of the structure strongly depend on core/shell sizes and the strength of the external magnetic field. We found that the maximum probability of finding the electron occurs in either the core or the shell region, depending on the value of the threshold core radius. This threshold radius separates two regions where physical behaviors undergo changes and the applied magnetic field acts as an additional confinement.
Collapse
Affiliation(s)
- Mohamed El-Yadri
- Group of Optoelectronic of Semiconductors and Nanomaterials, ENSAM, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Jawad El Hamdaoui
- Group of Optoelectronic of Semiconductors and Nanomaterials, ENSAM, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Noreddine Aghoutane
- Instituto de Alta Investigación, CEDENNA, Universidad de Tarapacá, Casilla 7D, Arica 1000000, Chile
| | - Laura M. Pérez
- Departamento de Física, FACI, Universidad de Tarapacá, Casilla 7D, Arica 1000000, Chile
| | - Sotirios Baskoutas
- Department of Materials Science, University of Patras, GR-26504 Patras, Greece
| | - David Laroze
- Instituto de Alta Investigación, CEDENNA, Universidad de Tarapacá, Casilla 7D, Arica 1000000, Chile
| | - Pablo Díaz
- Departamento de Ciencias Físicas, Universidad de La Frontera, Casilla 54-D, Temuco 4780000, Chile
| | - El Mustapha Feddi
- Group of Optoelectronic of Semiconductors and Nanomaterials, ENSAM, Mohammed V University in Rabat, Rabat 10100, Morocco
- Institute of Applied Physics, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid Ben Guerir, Ben Guerir 43150, Morocco
| |
Collapse
|
25
|
Segawa TF, Igarashi R. Nanoscale quantum sensing with Nitrogen-Vacancy centers in nanodiamonds - A magnetic resonance perspective. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2023; 134-135:20-38. [PMID: 37321756 DOI: 10.1016/j.pnmrs.2022.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
Nanodiamonds containing fluorescent Nitrogen-Vacancy (NV) centers are the smallest single particles, of which a magnetic resonance spectrum can be recorded at room temperature using optically-detected magnetic resonance (ODMR). By recording spectral shift or changes in relaxation rates, various physical and chemical quantities can be measured such as the magnetic field, orientation, temperature, radical concentration, pH or even NMR. This turns NV-nanodiamonds into nanoscale quantum sensors, which can be read out by a sensitive fluorescence microscope equipped with an additional magnetic resonance upgrade. In this review, we introduce the field of ODMR spectroscopy of NV-nanodiamonds and how it can be used to sense different quantities. Thereby we highlight both, the pioneering contributions and the latest results (covered until 2021) with a focus on biological applications.
Collapse
Affiliation(s)
- Takuya F Segawa
- Laboratory of Physical Chemistry, ETH Zurich, 8093 Zurich, Switzerland; Laboratory for Solid State Physics, ETH Zurich, 8093 Zurich, Switzerland.
| | - Ryuji Igarashi
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, 4-9-1, Anagawa, Inage-Ku, Chiba 263-8555, Japan; Takasaki Advanced Radiation Research Institute, National Institutes for Quantum Science and Technology, 1233 Watanuki, Takasaki, Gunma 370-1292, Japan; JST, PRESTO, Kawaguchi, Japan.
| |
Collapse
|
26
|
Wang X, Liu S, Zhang W, Peng H, Zhang M, Li Y, Guo Q, Wang W, Huang N, Liu L, Liu D. Silicon nanowire array overcomes chemotherapeutic resistance by inducing the differentiation of breast cancer stem cells. J Biomed Mater Res B Appl Biomater 2023. [PMID: 36929288 DOI: 10.1002/jbm.b.35249] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/18/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
Currently, traditional cancer treatment strategies are greatly challenged by the existence of cancer stem cells (CSCs), which are root cause of chemotherapy resistance. Differentiation therapy presents a novel therapeutic strategy for CSC-targeted therapy. However, there are very few studies on the induction of CSCs differentiation so far. Silicon nanowire array (SiNWA) with many unique properties is considered to be an excellent material for various applications ranging from biotechnology to biomedical applications. In this study, we report the SiNWA differentiates MCF-7-derived breast CSCs (BCSCs) into non-CSCs by modulating the morphology of cells. In vitro, the differentiated BCSCs lose the stemness properties and thus become sensitive to chemotherapeutic drugs, eventually leading to the death of BCSCs. Therefore, this work suggests a potential approach for overcoming chemotherapeutic resistance.
Collapse
Affiliation(s)
- Xiaotong Wang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Sisi Liu
- Cheng'an County Hospital of Traditional Chinese Medicine, Handan, People's Republic of China
| | - Wei Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Haotong Peng
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Miao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Yaping Li
- College of Public Health, Hebei University, Baoding, People's Republic of China
| | - Qi Guo
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Wenjing Wang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - Na Huang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| | - LiYan Liu
- Medical Comprehensive Experimental Centrer, Hebei University, Baoding, People's Republic of China
| | - Dandan Liu
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, People's Republic of China
| |
Collapse
|
27
|
Wong DCP, Ding JL. The mechanobiology of NK cells- 'Forcing NK to Sense' target cells. Biochim Biophys Acta Rev Cancer 2023; 1878:188860. [PMID: 36791921 DOI: 10.1016/j.bbcan.2023.188860] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/16/2023]
Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and kill cancer and infected cells, which makes them unique 'off-the-shelf' candidates for a new generation of immunotherapies. Biomechanical forces in homeostasis and pathophysiology accrue additional immune regulation for NK immune responses. Indeed, cellular and tissue biomechanics impact NK receptor clustering, cytoskeleton remodeling, NK transmigration through endothelial cells, nuclear mechanics, and even NK-dendritic cell interaction, offering a plethora of unexplored yet important dynamic regulation for NK immunotherapy. Such events are made more complex by the heterogeneity of human NK cells. A significant question remains on whether and how biochemical and biomechanical cues collaborate for NK cell mechanotransduction, a process whereby mechanical force is sensed, transduced, and translated to downstream mechanical and biochemical signalling. Herein, we review recent advances in understanding how NK cells perceive and mechanotransduce biophysical cues. We focus on how the cellular cytoskeleton crosstalk regulates NK cell function while bearing in mind the heterogeneity of NK cells, the direct and indirect mechanical cues for NK anti-tumor activity, and finally, engineering advances that are of translational relevance to NK cell biology at the systems level.
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Department of Biological Sciences, National University of Singapore, 117543, Singapore.
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, 117543, Singapore; Integrative Sciences and Engineering Programme, National University of Singapore, 119077, Singapore.
| |
Collapse
|
28
|
Ghani MW, Iqbal A, Ghani H, Bibi S, Wang Z, Pei R. Recent advances in nanocomposite-based delivery systems for targeted CRISPR/Cas delivery and therapeutic genetic manipulation. J Mater Chem B 2023. [PMID: 36779580 DOI: 10.1039/d2tb02610d] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
CRISPR/Cas systems are novel gene editing tools with tremendous capacity and accuracy for gene editing and hold great potential for therapeutic genetic manipulation. However, the lack of safe and efficient delivery methods for CRISPR/Cas and its guide RNA hinders their wide adoption for therapeutic applications. To this end, there is an increasing demand for safe, efficient, precise, and non-pathogenic delivery approaches, both in vitro and in vivo. With the convergence of nanotechnology and biomedicine, functional nanocomposites have demonstrated unparalleled sophistication to overcome the limits of CRISPR/Cas delivery. The tunability of the physicochemical properties of nanocomposites makes it very easy to conjugate them with different functional substances. The combinatorial application of diverse functional materials in the form of nanocomposites has shown excellent properties for CRISPR/Cas delivery at the target site with therapeutic potential. The recent highlights of selective organ targeting and phase I clinical trials for gene manipulation by CRISPR/Cas after delivery through LNPs are at the brink of making it to routine clinical practice. Here we summarize the recent advances in delivering CRISPR/Cas systems through nanocomposites for targeted delivery and therapeutic genome editing.
Collapse
Affiliation(s)
- Muhammad Waseem Ghani
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, P. R. China.,CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Science, Suzhou 215123, P. R. China.
| | - Ambreen Iqbal
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, P. R. China.,CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Science, Suzhou 215123, P. R. China.
| | - Hammad Ghani
- Basic Health Unit Laleka, Primary and Secondary Healthcare Department, Bahawalngar, 62300, Punjab, Pakistan
| | - Sidra Bibi
- Department of Biology, The Islamia University of Bahawalpur, Bahawalnagar Campus 62300, Pakistan
| | - Zixun Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, P. R. China.,CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Science, Suzhou 215123, P. R. China.
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, P. R. China.,CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Science, Suzhou 215123, P. R. China.
| |
Collapse
|
29
|
Wang J, Li P, Wang N, Wang J, Xing D. Antibacterial features of material surface: strong enough to serve as antibiotics? J Mater Chem B 2023; 11:280-302. [PMID: 36533438 DOI: 10.1039/d2tb02139k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacteria are small but need big efforts to control. The use of antibiotics not only produces superbugs that are increasingly difficult to inactivate, but also raises environmental concerns with the growing consumption. It is now believed that the antibacterial task can count on some physiochemical features of material surfaces, which can be anti-adhesive or bactericidal without releasing toxicants. It is necessary to evaluate to what extent can we rely on the surface design since the actual application scenarios will need the antibacterial performance to be sharp, robust, environmentally friendly, and long-lasting. Herein, we review the recent laboratory advances that have been classified based on the specific surface features, including hydrophobicity, charge potential, micromorphology, stiffness and viscosity, and photoactivity, and the antibacterial mechanisms of each feature are included to provide a basic rationale for future design. The significance of anti-biofilms is also introduced, given the big role of biofilms in bacteria-caused damage. A perspective on the potential wide application of antibacterial surface features as a substitute or supplement to antibiotics is then discussed. Surface design is no doubt a solution worthy to explore, and future success will be a result of further progress in multiple directions, including mechanism study and material preparation.
Collapse
Affiliation(s)
- Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China. .,CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, China Academy of Sciences, Qingdao 266071, China.
| | - Ping Li
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Ning Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, China Academy of Sciences, Qingdao 266071, China.
| | - Jing Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Institute of Oceanology, China Academy of Sciences, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
30
|
Chakrabarty P, Illath K, Kar S, Nagai M, Santra TS. Combinatorial physical methods for cellular therapy: Towards the future of cellular analysis? J Control Release 2023; 353:1084-1095. [PMID: 36538949 DOI: 10.1016/j.jconrel.2022.12.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
The physical energy activated techniques for cellular delivery and analysis is one of the most rapidly expanding research areas for a variety of biological and biomedical discoveries. These methods, such as electroporation, optoporation, sonoporation, mechanoporation, magnetoporation, etc., have been widely used in delivering different biomolecules into a range of primary and patient-derived cell types. However, the techniques when used individually have had limitations in delivery and co-delivery of diverse biomolecules in various cell types. In recent years, a number of studies have been performed by combining the different membrane disruption techniques, either sequentially or simultaneously, in a single study. The studies, referred to as combinatorial, or hybrid techniques, have demonstrated enhanced transfection, such as efficient macromolecular and gene delivery and co-delivery, at lower delivery parameters and with high cell viability. Such studies can open up new and exciting avenues for understanding the subcellular structure and consequently facilitate the development of novel therapeutic strategies. This review consequently aims at summarising the different developments in hybrid therapeutic techniques. The different methods discussed include mechano-electroporation, electro-sonoporation, magneto-mechanoporation, magnetic nanoparticles enhanced electroporation, and magnetic hyperthermia studies. We discuss the clinical status of the different methods and conclude with a discussion on the future prospects of the combinatorial techniques for cellular therapy and diagnostics.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Srabani Kar
- Department of Physics, Indian Institute of Science Education and Research, Tirupati, India
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India.
| |
Collapse
|
31
|
Wang Y, Wang N, Yang Y, Chen Y, Zhang Z. Cellular nanomechanics derived from pattern-dependent focal adhesion and cytoskeleton to balance gene transfection of malignant osteosarcoma. J Nanobiotechnology 2022; 20:499. [DOI: 10.1186/s12951-022-01713-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
AbstractGene transfection was supposed to be the most promising technology to overcome the vast majority of diseases and it has been popularly reported in clinical applications of gene therapy. In spite of the rapid development of novel transfection materials and methods, the influence of morphology-dependent nanomechanics of malignant osteosarcoma on gene transfection is still unsettled. In this study, cell spreading and adhesion area was adjusted by the prepared micropatterns to regulate focal adhesion (FA) formation and cytoskeletal organization in osteosarcoma cells. The micropattern-dependent FA and cytoskeleton could induce different cellular nanomechanics to affect cell functions. Our results indicated that transfection efficiency was improved with enlarging FA area and cell nanomechanics in micropatterned osteosarcoma. The difference of gene transfection in micropatterned cells was vigorously supported by cellular internalization capacity, Ki67 proliferation ability and YAP mechanotranduction through the regulation of focal adhesion and cytoskeletal mechanics. This study is an attempt to disclose the relationship of cell nanomechanics and gene transfection for efficient gene delivery and develop multifunctional nanomedicine biomaterials for accurate gene therapy in osteosarcoma cells.
Collapse
|
32
|
Maurizi E, Martella DA, Schiroli D, Merra A, Mustfa SA, Pellegrini G, Macaluso C, Chiappini C. Nanoneedles Induce Targeted siRNA Silencing of p16 in the Human Corneal Endothelium. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203257. [PMID: 36253148 PMCID: PMC9685449 DOI: 10.1002/advs.202203257] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/08/2022] [Indexed: 06/16/2023]
Abstract
Nanoneedles can target nucleic acid transfection to primary cells at tissue interfaces with high efficiency and minimal perturbation. The corneal endothelium is an ideal target for nanoneedle-mediated RNA interference therapy aimed at enhancing its proliferative capacity, necessary for tissue regeneration. This work develops a strategy for siRNA nanoninjection to the human corneal endothelium. Nanoneedles can deliver p16-targeting siRNA to primary human corneal endothelial cells in vitro without toxicity. The nanoinjection of siRNA induces p16 silencing and increases cell proliferation, as monitored by ki67 expression. Furthermore, siRNA nanoinjection targeting the human corneal endothelium is nontoxic ex vivo, and silences p16 in transfected cells. These data indicate that nanoinjection can support targeted RNA interference therapy for the treatment of endothelial corneal dysfunction.
Collapse
Affiliation(s)
- Eleonora Maurizi
- Dentistry Centre LabUniversity of Parmavia Gramsci 14Parma43126Italy
- Centre for Regenerative Medicine ‘‘S. Ferrari’’University of Modena and Reggio EmiliaModena41125Italy
| | | | - Davide Schiroli
- Transfusion Medicine UnitAzienda USL‐IRCCSReggio Emilia42122Italy
| | | | - Salman Ahmad Mustfa
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- AstraZenecaGranta Park, Great AbingtonCambridgeCB21 6GHUnited Kingdom
| | - Graziella Pellegrini
- Centre for Regenerative Medicine ‘‘S. Ferrari’’University of Modena and Reggio EmiliaModena41125Italy
- Holostem Terapie Avanzate S.r.l.Modena41125Italy
| | - Claudio Macaluso
- Dentistry Centre LabUniversity of Parmavia Gramsci 14Parma43126Italy
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonSE1 9RTUK
- London Centre for NanotechnologyKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
33
|
Zeng Y, Zhuang Y, Vinod B, Guo X, Mitra A, Chen P, Saggio I, Shivashankar GV, Gao W, Zhao W. Guiding Irregular Nuclear Morphology on Nanopillar Arrays for Malignancy Differentiation in Tumor Cells. NANO LETTERS 2022; 22:7724-7733. [PMID: 35969027 DOI: 10.1021/acs.nanolett.2c01849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
For more than a century, abnormal nuclei in tumor cells, presenting subnuclear invaginations and folds on the nuclear envelope, have been known to be associated with high malignancy and poor prognosis. However, current nuclear morphology analysis focuses on the features of the entire nucleus, overlooking the malignancy-related subnuclear features in nanometer scale. The main technical challenge is to probe such tiny and randomly distributed features inside cells. We here employ nanopillar arrays to guide subnuclear features into ordered patterns, enabling their quantification as a strong indicator of cell malignancy. Both breast and liver cancer cells were validated as well as the quantification of nuclear abnormality heterogeneity. The alterations of subnuclear patterns were also explored as effective readouts for drug treatment. We envision that this nanopillar-enabled quantification of subnuclear abnormal features in tumor cells opens a new angle in characterizing malignant cells and studying the unique nuclear biology in cancer.
Collapse
Affiliation(s)
- Yongpeng Zeng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
| | - Yinyin Zhuang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
| | - Benjamin Vinod
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
| | - Xiangfu Guo
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
| | - Aninda Mitra
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie Charles Darwin, Sapienza Università di Roma, 00185, Roma, Italy
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
- CNR Institute of Molecular Biology and Pathology, 00185, Rome, Italy
| | - G V Shivashankar
- Department of Health Sciences & Technology (D-HEST), ETH Zurich, 8093, Zurich, Switzerland
- Paul Scherrer Institute, 5232, Villigen, Switzerland
| | - Weibo Gao
- Division of Physics and Applied Physics, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
- The Photonics Institute and Centre for Disruptive Photonic Technologies, Nanyang Technological University, 637371, Singapore
| | - Wenting Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| |
Collapse
|
34
|
Fang J, Huang S, Liu F, He G, Li X, Huang X, Chen HJ, Xie X. Semi-Implantable Bioelectronics. NANO-MICRO LETTERS 2022; 14:125. [PMID: 35633391 PMCID: PMC9148344 DOI: 10.1007/s40820-022-00818-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 06/15/2023]
Abstract
Developing techniques to effectively and real-time monitor and regulate the interior environment of biological objects is significantly important for many biomedical engineering and scientific applications, including drug delivery, electrophysiological recording and regulation of intracellular activities. Semi-implantable bioelectronics is currently a hot spot in biomedical engineering research area, because it not only meets the increasing technical demands for precise detection or regulation of biological activities, but also provides a desirable platform for externally incorporating complex functionalities and electronic integration. Although there is less definition and summary to distinguish it from the well-reviewed non-invasive bioelectronics and fully implantable bioelectronics, semi-implantable bioelectronics have emerged as highly unique technology to boost the development of biochips and smart wearable device. Here, we reviewed the recent progress in this field and raised the concept of "Semi-implantable bioelectronics", summarizing the principle and strategies of semi-implantable device for cell applications and in vivo applications, discussing the typical methodologies to access to intracellular environment or in vivo environment, biosafety aspects and typical applications. This review is meaningful for understanding in-depth the design principles, materials fabrication techniques, device integration processes, cell/tissue penetration methodologies, biosafety aspects, and applications strategies that are essential to the development of future minimally invasive bioelectronics.
Collapse
Affiliation(s)
- Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Shuang Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Fanmao Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
35
|
Sun S, Yang Y, Niu H, Luo M, Wu ZS. Design and application of DNA nanostructures for organelle-targeted delivery of anticancer drugs. Expert Opin Drug Deliv 2022; 19:707-723. [PMID: 35618266 DOI: 10.1080/17425247.2022.2083603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION DNA nanostructures targeting organelles are of great significance for the early diagnosis and precise therapy of human cancers. This review is expected to promote the development of DNA nanostructure-based cancer treatment with organelle-level precision in the future. AREAS COVERED In this review, we introduce the different principles for targeting organelles, summarize the progresses in the development of organelle-targeting DNA nanostructures, highlight their advantages and applications in disease treatment, and discuss current challenges and future prospects. EXPERT OPINION Accurate targeting is a basic problem for effective cancer treatment. However, current DNA nanostructures cannot meet the actual needs. Targeting specific organelles is expected to further improve the therapeutic effect and overcome tumor cell resistance, thereby holding great practical significance for tumor treatment in the clinic. With the deepening of the research on the molecular mechanism of disease development, especially on tumorigenesis and tumor progression, and increasing understanding of the behavior of biological materials in living cells, more versatile DNA nanostructures will be constructed to target subcellular organelles for drug delivery, essentially promoting the early diagnosis of cancers, classification, precise therapy and the estimation of prognosis in the future.
Collapse
Affiliation(s)
- Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China.,Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Huimin Niu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China.,Fujian Key Laboratory of Aptamers Technology, The 900th Hospital of Joint Logistics Support Force, Fuzhou 350025, China
| | - Mengxue Luo
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| |
Collapse
|
36
|
Topography-Mediated Enhancement of Nonviral Gene Delivery in Stem Cells. Pharmaceutics 2022; 14:pharmaceutics14051096. [PMID: 35631682 PMCID: PMC9142897 DOI: 10.3390/pharmaceutics14051096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/09/2022] [Accepted: 05/18/2022] [Indexed: 02/07/2023] Open
Abstract
Gene delivery holds great promise for bioengineering, biomedical applications, biosensors, diagnoses, and gene therapy. In particular, the influence of topography on gene delivery is considered to be an attractive approach due to low toxicity and localized delivery properties. Even though many gene vectors and transfection systems have been developed to enhance transfection potential and combining it with other forms of stimulations could even further enhance it. Topography is an interesting surface property that has been shown to stimulate differentiation, migration, cell morphology, and cell mechanics. Therefore, it is envisioned that topography might also be able to stimulate transfection. In this study, we tested the hypothesis “topography is able to regulate transfection efficiency”, for which we used nano- and microwave-like topographical substrates with wavelengths ranging from 500 nm to 25 µm and assessed the transfectability of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and myoblasts. For transfection, Lipofectamine 2000 and a gene encoding plasmid for red-fluorescent protein (m-Cherry) were used and topography-induced cell morphology and transfection efficiency was analyzed. As a result, topography directs cell spreading, elongation, and proliferation as well as the transfection efficiency, which were investigated but were found not to be correlated and dependent on the cell type. A 55% percent improvement of transfection efficiency was identified for hBM-MSCs grown on 2 µm wrinkles (24.3%) as compared to hBM-MSCs cultured on flat controls (15.7%). For myoblast cells, the highest gene-expression efficiency (46.1%) was observed on the 10 µm topography, which enhanced the transfection efficiency by 64% as compared to the flat control (28.1%). From a qualitative assessment, it was observed that the uptake capacity of cationic complexes of TAMRA-labeled oligodeoxynucleotides (ODNs) was not topography-dependent but that the intracellular release was faster, as indicated by the positively stained nuclei on 2 μm for hBM-MSCs and 10 μm for myoblasts. The presented results indicate that topography enhances the gene-delivery capacity and that the responses are dependent on cell type. This study demonstrates the important role of topography on cell stimulation for gene delivery as well as understanding the uptake capacity of lipoplexes and may be useful for developing advanced nonviral gene delivery strategies.
Collapse
|
37
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
38
|
Shakoor A, Gao W, Zhao L, Jiang Z, Sun D. Advanced tools and methods for single-cell surgery. MICROSYSTEMS & NANOENGINEERING 2022; 8:47. [PMID: 35502330 PMCID: PMC9054775 DOI: 10.1038/s41378-022-00376-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 06/14/2023]
Abstract
Highly precise micromanipulation tools that can manipulate and interrogate cell organelles and components must be developed to support the rapid development of new cell-based medical therapies, thereby facilitating in-depth understanding of cell dynamics, cell component functions, and disease mechanisms. This paper presents a literature review on micro/nanomanipulation tools and their control methods for single-cell surgery. Micromanipulation methods specifically based on laser, microneedle, and untethered micro/nanotools are presented in detail. The limitations of these techniques are also discussed. The biological significance and clinical applications of single-cell surgery are also addressed in this paper.
Collapse
Affiliation(s)
- Adnan Shakoor
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Wendi Gao
- State Key Laboratory for Manufacturing Systems Engineering, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technologies, The School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Libo Zhao
- State Key Laboratory for Manufacturing Systems Engineering, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technologies, The School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Zhuangde Jiang
- State Key Laboratory for Manufacturing Systems Engineering, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technologies, The School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Dong Sun
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- State Key Laboratory for Manufacturing Systems Engineering, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technologies, The School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
39
|
Zhang A, Fang J, Li X, Wang J, Chen M, Chen HJ, He G, Xie X. Cellular nanointerface of vertical nanostructure arrays and its applications. NANOSCALE ADVANCES 2022; 4:1844-1867. [PMID: 36133409 PMCID: PMC9419580 DOI: 10.1039/d1na00775k] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/28/2021] [Indexed: 06/16/2023]
Abstract
Vertically standing nanostructures with various morphologies have been developed with the emergence of the micro-/nanofabrication technology. When cells are cultured on them, various bio-nano interfaces between cells and vertical nanostructures would impact the cellular activities, depending on the shape, density, and height of nanostructures. Many cellular pathway activation processes involving a series of intracellular molecules (proteins, RNA, DNA, enzymes, etc.) would be triggered by the cell morphological changes induced by nanostructures, affecting the cell proliferation, apoptosis, differentiation, immune activation, cell adhesion, cell migration, and other behaviors. In addition, the highly localized cellular nanointerface enhances coupled stimulation on cells. Therefore, understanding the mechanism of the cellular nanointerface can not only provide innovative tools for regulating specific cell functions but also offers new aspects to understand the fundamental cellular activities that could facilitate the precise monitoring and treatment of diseases in the future. This review mainly describes the fabrication technology of vertical nanostructures, analyzing the formation of cellular nanointerfaces and the effects of cellular nanointerfaces on cells' fates and functions. At last, the applications of cellular nanointerfaces based on various nanostructures are summarized.
Collapse
Affiliation(s)
- Aihua Zhang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- School of Biomedical Engineering, Sun Yat-Sen University Guangzhou 510006 China
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| | - Meiwan Chen
- Institute of Chinese Medical Sciences, University of Macau Taipa Macau SAR China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University Guangzhou 511436 P. R. China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University Guangzhou 510006 Guangdong Province China
- The First Affiliated Hospital of Sun Yat-Sen University Guangzhou 510080 China
| |
Collapse
|
40
|
Numano R, Goryu A, Kubota Y, Sawahata H, Yamagiwa S, Matsuo M, Iimura T, Tei H, Ishida M, Kawano T. Nanoscale-tipped wire array injections transfer DNA directly into brain cells ex vivo and in vivo. FEBS Open Bio 2022; 12:835-851. [PMID: 35293154 PMCID: PMC8972050 DOI: 10.1002/2211-5463.13377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/24/2021] [Accepted: 02/04/2022] [Indexed: 11/26/2022] Open
Abstract
Genetic modification to restore cell functions in the brain can be performed through the delivery of biomolecules in a minimally invasive manner into live neuronal cells within brain tissues. However, conventional nanoscale needles are too short (lengths of ~10 µm) to target neuronal cells in ~1‐mm‐thick brain tissues because the neuronal cells are located deep within the tissue. Here, we report the use of nanoscale‐tipped wire (NTW) arrays with diameters < 100 nm and wire lengths of ~200 µm to address biomolecule delivery issues. The NTW arrays were manufactured by growth of silicon microwire arrays and nanotip formation. This technique uses pinpoint, multiple‐cell DNA injections in deep areas of brain tissues, enabling target cells to be marked by fluorescent protein (FP) expression vectors. This technique has potential for use for electrophysiological recordings and biological transfection into neuronal cells. Herein, simply pressing an NTW array delivers and expresses plasmid DNA in multiple‐cultured cells and multiple‐neuronal cells within a brain slice with reduced cell damage. Additionally, DNA transfection is demonstrated using brain cells ex vivo and in vivo. Moreover, knockdown of a critical clock gene after injecting a short hairpin RNA (shRNA) and a genome‐editing vector demonstrates the potential to genetically alter the function of living brain cells, for example, pacemaker cells of the mammalian circadian rhythms. Overall, our NTW array injection technique enables genetic and functional modification of living cells in deep brain tissue areas, both ex vivo and in vivo.
Collapse
Affiliation(s)
- Rika Numano
- Department of Applied Chemistry and Life Science, Toyohashi University of Technology, Japan.,Electronics-Inspired Interdisciplinary Research Institute (EIIRIS), Toyohashi University of Technology, Japan
| | - Akihiro Goryu
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan
| | - Yoshihiro Kubota
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan
| | - Hirohito Sawahata
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan.,National Institute of Technology, Ibaraki College, Japan
| | - Shota Yamagiwa
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan
| | - Minako Matsuo
- Department of Applied Chemistry and Life Science, Toyohashi University of Technology, Japan.,Electronics-Inspired Interdisciplinary Research Institute (EIIRIS), Toyohashi University of Technology, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Hajime Tei
- Graduate School of Natural Science and Technology, Kanazawa University, Japan
| | - Makoto Ishida
- Electronics-Inspired Interdisciplinary Research Institute (EIIRIS), Toyohashi University of Technology, Japan.,Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan
| | - Takeshi Kawano
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Japan
| |
Collapse
|
41
|
Mariano A, Lubrano C, Bruno U, Ausilio C, Dinger NB, Santoro F. Advances in Cell-Conductive Polymer Biointerfaces and Role of the Plasma Membrane. Chem Rev 2022; 122:4552-4580. [PMID: 34582168 PMCID: PMC8874911 DOI: 10.1021/acs.chemrev.1c00363] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Indexed: 02/07/2023]
Abstract
The plasma membrane (PM) is often described as a wall, a physical barrier separating the cell cytoplasm from the extracellular matrix (ECM). Yet, this wall is a highly dynamic structure that can stretch, bend, and bud, allowing cells to respond and adapt to their surrounding environment. Inspired by shapes and geometries found in the biological world and exploiting the intrinsic properties of conductive polymers (CPs), several biomimetic strategies based on substrate dimensionality have been tailored in order to optimize the cell-chip coupling. Furthermore, device biofunctionalization through the use of ECM proteins or lipid bilayers have proven successful approaches to further maximize interfacial interactions. As the bio-electronic field aims at narrowing the gap between the electronic and the biological world, the possibility of effectively disguising conductive materials to "trick" cells to recognize artificial devices as part of their biological environment is a promising approach on the road to the seamless platform integration with cells.
Collapse
Affiliation(s)
- Anna Mariano
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Claudia Lubrano
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Ugo Bruno
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Chiara Ausilio
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Nikita Bhupesh Dinger
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Dipartimento
di Chimica, Materiali e Produzione Industriale, Università di Napoli Federico II, 80125 Naples, Italy
| | - Francesca Santoro
- Tissue
Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
42
|
Biocompatible surface functionalization architecture for a diamond quantum sensor. Proc Natl Acad Sci U S A 2022; 119:2114186119. [PMID: 35193961 PMCID: PMC8872777 DOI: 10.1073/pnas.2114186119] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2022] [Indexed: 01/02/2023] Open
Abstract
Diamond-based quantum sensing enables nanoscale measurements of biological systems with unprecedented sensitivity. Potential applications of this emerging technology range from the investigation of fundamental biological processes to the development of next-generation medical diagnostics devices. One of the main challenges faced by bioquantum sensing is the need to interface quantum sensors with biological target systems. Specifically, such an interface needs to maintain the highly fragile quantum states of our sensor and at the same time be able to fish intact biomolecules out of solution and immobilize them on our quantum sensor surface. Our work overcomes these challenges by combining tools from quantum engineering, single-molecule biophysics, and material processing. Quantum metrology enables some of the most precise measurements. In the life sciences, diamond-based quantum sensing has led to a new class of biophysical sensors and diagnostic devices that are being investigated as a platform for cancer screening and ultrasensitive immunoassays. However, a broader application in the life sciences based on nanoscale NMR spectroscopy has been hampered by the need to interface highly sensitive quantum bit (qubit) sensors with their biological targets. Here, we demonstrate an approach that combines quantum engineering with single-molecule biophysics to immobilize individual proteins and DNA molecules on the surface of a bulk diamond crystal that hosts coherent nitrogen vacancy qubit sensors. Our thin (sub–5 nm) functionalization architecture provides precise control over the biomolecule adsorption density and results in near-surface qubit coherence approaching 100 μs. The developed architecture remains chemically stable under physiological conditions for over 5 d, making our technique compatible with most biophysical and biomedical applications.
Collapse
|
43
|
Mu H, Zeng Y, Zhuang Y, Gao W, Zhou Y, Rajalingam K, Zhao W. Patterning of Oncogenic Ras Clustering in Live Cells Using Vertically Aligned Nanostructure Arrays. NANO LETTERS 2022; 22:1007-1016. [PMID: 35044178 DOI: 10.1021/acs.nanolett.1c03886] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
As a dominant oncogenic protein, Ras is well-known to segregate into clusters on the plasma membrane for activating downstream signaling. However, current technologies for direct measurements of Ras clustering are limited to sophisticated high-resolution techniques like electron microscopy and fluorescence lifetime imaging. To further promote fundamental investigations and the related drug development, we hereby introduce a nanobar-based platform which effectively guides Ras clusters into quantifiable patterns in live cells that is resolvable under conventional microscopy. Major Ras isoforms, K-Ras, H-Ras, and N-Ras, were differentiated, as well as their highly prevalent oncogenic mutants G12V and G13D. Moreover, the isoform specificity and the sensitivity of a Ras inhibitor were successfully characterized on nanobars. We envision that this nanobar-based platform will serve as an effective tool to read Ras clustering on the plasma membrane, enabling a novel avenue both to decipher Ras regulations and to facilitate anti-Ras drug development.
Collapse
Affiliation(s)
- Huanwen Mu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
- Ageing Research Institute for Society and Education, Nanyang Technological University, Singapore 637335, Singapore
| | - Yongpeng Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Yinyin Zhuang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Weibo Gao
- Division of Physics and Applied Physics, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
- The Photonics Institute and Centre for Disruptive Photonic Technologies, Nanyang Technological University, Singapore 637371, Singapore
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030, United States
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center and UTHealth Graduate School of Biomedical Sciences, Houston, Texas 77030, United States
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
- University Cancer Center Mainz, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| |
Collapse
|
44
|
Sayed N, Allawadhi P, Khurana A, Singh V, Navik U, Pasumarthi SK, Khurana I, Banothu AK, Weiskirchen R, Bharani KK. Gene therapy: Comprehensive overview and therapeutic applications. Life Sci 2022; 294:120375. [PMID: 35123997 DOI: 10.1016/j.lfs.2022.120375] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023]
Abstract
Gene therapy is the product of man's quest to eliminate diseases. Gene therapy has three facets namely, gene silencing using siRNA, shRNA and miRNA, gene replacement where the desired gene in the form of plasmids and viral vectors, are directly administered and finally gene editing based therapy where mutations are modified using specific nucleases such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regulatory interspaced short tandem repeats (CRISPR)/CRISPR-associated protein (Cas)-associated nucleases. Transfer of gene is either through transformation where under specific conditions the gene is directly taken up by the bacterial cells, transduction where a bacteriophage is used to transfer the genetic material and lastly transfection that involves forceful delivery of gene using either viral or non-viral vectors. The non-viral transfection methods are subdivided into physical, chemical and biological. The physical methods include electroporation, biolistic, microinjection, laser, elevated temperature, ultrasound and hydrodynamic gene transfer. The chemical methods utilize calcium- phosphate, DAE-dextran, liposomes and nanoparticles for transfection. The biological methods are increasingly using viruses for gene transfer, these viruses could either integrate within the genome of the host cell conferring a stable gene expression, whereas few other non-integrating viruses are episomal and their expression is diluted proportional to the cell division. So far, gene therapy has been wielded in a plethora of diseases. However, coherent and innocuous delivery of genes is among the major hurdles in the use of this promising therapy. Hence this review aims to highlight the current options available for gene transfer along with the advantages and limitations of every method.
Collapse
Affiliation(s)
- Nilofer Sayed
- Department of Pharmacy, Pravara Rural Education Society's (P.R.E.S.'s) College of Pharmacy, Shreemati Nathibai Damodar Thackersey (SNDT) Women's University, Nashik 400020, Maharashtra, India
| | - Prince Allawadhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT), Roorkee, Roorkee, Uttarakhand 247667, India
| | - Amit Khurana
- Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi 110016, India; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India; Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Mamnoor, Warangal 506166, Telangana, India; Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Vishakha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT), Roorkee, Roorkee, Uttarakhand 247667, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | | | - Isha Khurana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Anil Kumar Banothu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, Pauwelsstr. 30, D-52074 Aachen, Germany.
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), PVNRTVU, Mamnoor, Warangal 506166, Telangana, India.
| |
Collapse
|
45
|
Loh DM, Nava M, Nocera DG. Polypyrrole-Silicon Nanowire Arrays for Controlled Intracellular Cargo Delivery. NANO LETTERS 2022; 22:366-371. [PMID: 34965139 DOI: 10.1021/acs.nanolett.1c04033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Intracellular cargo delivery is a critical and challenging step in controlling cell states. Silicon nanowire (NW) arrays have emerged as a powerful platform for accessing the intracellular space through a combination of their nanoscale dimensions and electrical properties. Here, we develop and characterize a conductive polypyrrole (PPy)-NW device for temporally controlled intracellular delivery. Fluorescent cargos, doped in electroresponsive PPy matrices at wire tips as well as entire NW arrays, are released with an applied reducing potential. Intracellular delivery into endothelial cells from PPy-Si substrates demonstrated comparable kinetics to solution-based delivery methods while requiring an order of magnitude less cargo loading. This hybrid polymer-semiconductor platform extends methods available for intracellular delivery and links electrical signaling from artificial systems with living molecular transduction.
Collapse
Affiliation(s)
- Daniel M Loh
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Matthew Nava
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Daniel G Nocera
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
46
|
Protasiuk L, Serov NS, Lokteva AV, Kladko D, Koshel EI, Vinogradov VV. Mechano-bactericidal anisotropic particles for oral biofilm treatment. J Mater Chem B 2022; 10:4867-4877. [DOI: 10.1039/d2tb00582d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bacterial biofilms stand for the main etiological factor of dental diseases worldwide. At present, toothpaste with bactericidal chemicals as well as abrasive materials are used as preventive care. However, chemicals...
Collapse
|
47
|
Chakrabarty P, Gupta P, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic mechanoporation for cellular delivery and analysis. Mater Today Bio 2022; 13:100193. [PMID: 35005598 PMCID: PMC8718663 DOI: 10.1016/j.mtbio.2021.100193] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Highly efficient intracellular delivery strategies are essential for developing therapeutic, diagnostic, biological, and various biomedical applications. The recent advancement of micro/nanotechnology has focused numerous researches towards developing microfluidic device-based strategies due to the associated high throughput delivery, cost-effectiveness, robustness, and biocompatible nature. The delivery strategies can be carrier-mediated or membrane disruption-based, where membrane disruption methods find popularity due to reduced toxicity, enhanced delivery efficiency, and cell viability. Among all of the membrane disruption techniques, the mechanoporation strategies are advantageous because of no external energy source required for membrane deformation, thereby achieving high delivery efficiencies and increased cell viability into different cell types with negligible toxicity. The past two decades have consequently seen a tremendous boost in mechanoporation-based research for intracellular delivery and cellular analysis. This article provides a brief review of the most recent developments on microfluidic-based mechanoporation strategies such as microinjection, nanoneedle arrays, cell-squeezing, and hydroporation techniques with their working principle, device fabrication, cellular delivery, and analysis. Moreover, a brief discussion of the different mechanoporation strategies integrated with other delivery methods has also been provided. Finally, the advantages, limitations, and future prospects of this technique are discussed compared to other intracellular delivery techniques.
Collapse
Affiliation(s)
- Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| | - Srabani Kar
- Department of Electrical Engineering, University of Cambridge, Cambridge, CB30FA, UK
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Aichi, Japan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
48
|
Choi S, Lee J, Lee K, Yoon SM, Yoon M. Porphyrin-decorated ZnO nanowires as nanoscopic injectors for phototheragnosis of cancer cells. NEW J CHEM 2022. [DOI: 10.1039/d2nj02084j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Newly synthesized protoporphyrin-decorated ZnO-nanowires exhibited optical waveguided and photodynamic properties to be useful nanoscopic injectors for photo-theragnosis of cancer cells.
Collapse
Affiliation(s)
- Sunyoung Choi
- Molecular/Nano Photochemistry and Photonics Lab, Department of Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jooran Lee
- Molecular/Nano Photochemistry and Photonics Lab, Department of Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
- TheraNovis Inc. 32 Seongnae-ro 6-gil, Gangdong-gu, Seoul, Republic of Korea
| | - Kangmin Lee
- Department of Chemistry, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, Republic of Korea
- Wonkwang Materials Institute of Science and Technology, 460 Iksandae-ro, Iksan, Jeonbuk, Republic of Korea
| | - Seok Min Yoon
- Department of Chemistry, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk, Republic of Korea
- Wonkwang Materials Institute of Science and Technology, 460 Iksandae-ro, Iksan, Jeonbuk, Republic of Korea
| | - Minjoong Yoon
- Molecular/Nano Photochemistry and Photonics Lab, Department of Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
49
|
Penedo M, Miyazawa K, Okano N, Furusho H, Ichikawa T, Alam MS, Miyata K, Nakamura C, Fukuma T. Visualizing intracellular nanostructures of living cells by nanoendoscopy-AFM. SCIENCE ADVANCES 2021; 7:eabj4990. [PMID: 34936434 PMCID: PMC10954033 DOI: 10.1126/sciadv.abj4990] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/10/2021] [Indexed: 06/14/2023]
Abstract
Atomic force microscopy (AFM) is the only technique that allows label-free imaging of nanoscale biomolecular dynamics, playing a crucial role in solving biological questions that cannot be addressed by other major bioimaging tools (fluorescence or electron microscopy). However, such imaging is possible only for systems either extracted from cells or reconstructed on solid substrates. Thus, nanodynamics inside living cells largely remain inaccessible with the current nanoimaging techniques. Here, we overcome this limitation by nanoendoscopy-AFM, where a needle-like nanoprobe is inserted into a living cell, presenting actin fiber three-dimensional (3D) maps, and 2D nanodynamics of the membrane inner scaffold, resulting in undetectable changes in cell viability. Unlike previous AFM methods, the nanoprobe directly accesses the target intracellular components, exploiting all the AFM capabilities, such as high-resolution imaging, nanomechanical mapping, and molecular recognition. These features should greatly expand the range of intracellular structures observable in living cells.
Collapse
Affiliation(s)
- Marcos Penedo
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Keisuke Miyazawa
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Division of Electric Engineering and Computer Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Naoko Okano
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Hirotoshi Furusho
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Takehiko Ichikawa
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Mohammad Shahidul Alam
- Division of Nano Life Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Kazuki Miyata
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Division of Electric Engineering and Computer Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Division of Nano Life Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Chikashi Nakamura
- AIST-INDIA Diverse Assets and Applications International Laboratory (DAILAB), Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8565, Japan
| | - Takeshi Fukuma
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
- Division of Electric Engineering and Computer Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Faculty of Frontier Engineering, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
- Division of Nano Life Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
50
|
High‐Efficient and Dosage‐Controllable Intracellular Cargo Delivery through Electrochemical Metal–Organic Hybrid Nanogates. SMALL SCIENCE 2021. [DOI: 10.1002/smsc.202100069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|