1
|
Xie J, Xiang J, Shen Y, Shao S. Mechanistic Insights into the Tools for Intracellular Protein Delivery. CHEM & BIO ENGINEERING 2025; 2:132-155. [PMID: 40171130 PMCID: PMC11955855 DOI: 10.1021/cbe.4c00168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 04/03/2025]
Abstract
Proteins are an important therapeutic modality in modern medicine. However, their inherent inability to traverse cell membranes essentially limits their application to extracellular targets. Recent advances in intracellular protein delivery have enabled access to traditionally "undruggable" intracellular targets and paved the way to precisely modulate cellular functions. This Review provides a comprehensive examination of the key mechanisms and emerging technologies that facilitate the transport of functional proteins across cellular membranes. Delivery methods are categorized into physical, chemical, and biological approaches, each with distinct advantages and limitations. Physical methods enable direct protein entry but often pose challenges related to invasiveness and technical complexity. Chemical strategies offer customizable solutions with enhanced control over cellular targeting and uptake, yet may face issues with cytotoxicity and scalability. Biological approaches leverage naturally occurring processes to achieve efficient intracellular transport, though regulatory and production consistency remain hurdles. By highlighting recent advancements, challenges, and opportunities within each approach, this review underscores the potential of intracellular protein delivery technologies to unlock new therapeutic pathways and transform drug development paradigms.
Collapse
Affiliation(s)
- Jingwen Xie
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Jiajia Xiang
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Youqing Shen
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| | - Shiqun Shao
- Zhejiang
Key Laboratory of Smart Biomaterials and Center for Bionanoengineering,
College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310058, China
| |
Collapse
|
2
|
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
|
3
|
Sanaee M, Ronquist KG, Sandberg E, Morrell JM, Widengren J, Gallo K. Antibody-Loading of Biological Nanocarrier Vesicles Derived from Red-Blood-Cell Membranes. ACS OMEGA 2024; 9:22711-22718. [PMID: 38826552 PMCID: PMC11137724 DOI: 10.1021/acsomega.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 06/04/2024]
Abstract
Antibodies, disruptive potent therapeutic agents against pharmacological targets, face a barrier in crossing immune systems and cellular membranes. To overcome these, various strategies have been explored including shuttling via liposomes or biocamouflaged nanoparticles. Here, we demonstrate the feasibility of loading antibodies into exosome-mimetic nanovesicles derived from human red-blood-cell membranes, which can act as nanocarriers for intracellular delivery. Goat-antichicken antibodies are loaded into erythrocyte-derived nanovesicles, and their loading yields are characterized and compared with smaller dUTP-cargo molecules. Applying dual-color coincident fluorescence burst analyses, the loading yield of nanocarriers is rigorously profiled at the single-vesicle level, overcoming challenges due to size-heterogeneity and demonstrating a maximum antibody-loading yield of 38-41% at the optimal vesicle radius of 52 nm. The achieved average loading yields, amounting to 14% across the entire nanovesicle population, with more than two antibodies per loaded vesicle, are fully comparable to those obtained for the much smaller dUTP molecules loaded in the nanovesicles after additional exosome-spin-column purification. The results suggest a promising new avenue for therapeutic delivery of antibodies, potentially encompassing also intracellular targets and suitable for large-scale pharmacological applications, which relies on the exosome-mimetic properties, biocompatibility, and low-immunogenicity of bioengineered nanocarriers synthesized from human erythrocyte membranes.
Collapse
Affiliation(s)
- Maryam Sanaee
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - K. Göran Ronquist
- Department
of Clinical Sciences, Swedish University
of Agricultural Sciences, Uppsala 75007, Sweden
| | - Elin Sandberg
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Jane M. Morrell
- Department
of Clinical Sciences, Swedish University
of Agricultural Sciences, Uppsala 75007, Sweden
| | - Jerker Widengren
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Katia Gallo
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| |
Collapse
|
4
|
Alegria AD, Joshi AS, Mendana JB, Khosla K, Smith KT, Auch B, Donovan M, Bischof J, Gohl DM, Kodandaramaiah SB. High-throughput genetic manipulation of multicellular organisms using a machine-vision guided embryonic microinjection robot. Genetics 2024; 226:iyae025. [PMID: 38373262 PMCID: PMC10990426 DOI: 10.1093/genetics/iyae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 02/21/2024] Open
Abstract
Microinjection is a technique used for transgenesis, mutagenesis, cell labeling, cryopreservation, and in vitro fertilization in multiple single and multicellular organisms. Microinjection requires specialized skills and involves rate-limiting and labor-intensive preparatory steps. Here, we constructed a machine-vision guided generalized robot that fully automates the process of microinjection in fruit fly (Drosophila melanogaster) and zebrafish (Danio rerio) embryos. The robot uses machine learning models trained to detect embryos in images of agar plates and identify specific anatomical locations within each embryo in 3D space using dual view microscopes. The robot then serially performs a microinjection in each detected embryo. We constructed and used three such robots to automatically microinject tens of thousands of Drosophila and zebrafish embryos. We systematically optimized robotic microinjection for each species and performed routine transgenesis with proficiency comparable to highly skilled human practitioners while achieving up to 4× increases in microinjection throughput in Drosophila. The robot was utilized to microinject pools of over 20,000 uniquely barcoded plasmids into 1,713 embryos in 2 days to rapidly generate more than 400 unique transgenic Drosophila lines. This experiment enabled a novel measurement of the number of independent germline integration events per successfully injected embryo. Finally, we showed that robotic microinjection of cryoprotective agents in zebrafish embryos significantly improves vitrification rates and survival of cryopreserved embryos post-thaw as compared to manual microinjection. We anticipate that the robot can be used to carry out microinjection for genome-wide manipulation and cryopreservation at scale in a wide range of organisms.
Collapse
Affiliation(s)
- Andrew D Alegria
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Amey S Joshi
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jorge Blanco Mendana
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kanav Khosla
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kieran T Smith
- Department of Fisheries, Wildlife and Conservation Biology, University of Minnesota, St. Paul, MN 55108, USA
| | - Benjamin Auch
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Margaret Donovan
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - John Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daryl M Gohl
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Suhasa B Kodandaramaiah
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
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
|
6
|
Omura M, Morimoto K, Araki Y, Hirose H, Kawaguchi Y, Kitayama Y, Goto Y, Harada A, Fujii I, Takatani-Nakase T, Futaki S, Nakase I. Inkjet-Based Intracellular Delivery System that Effectively Utilizes Cell-Penetrating Peptides for Cytosolic Introduction of Biomacromolecules through the Cell Membrane. ACS APPLIED MATERIALS & INTERFACES 2023; 15:47855-47865. [PMID: 37792057 PMCID: PMC10592309 DOI: 10.1021/acsami.3c01650] [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: 02/05/2023] [Accepted: 07/24/2023] [Indexed: 10/05/2023]
Abstract
In the drug delivery system, the cytosolic delivery of biofunctional molecules such as enzymes and genes must achieve sophisticated activities in cells, and microinjection and electroporation systems are typically used as experimental techniques. These methods are highly reliable, and they have high intracellular transduction efficacy. However, a high degree of proficiency is necessary, and induced cytotoxicity is considered as a technical problem. In this research, a new intracellular introduction technology was developed through the cell membrane using an inkjet device and cell-penetrating peptides (CPPs). Using the inkjet system, the droplet volume, droplet velocity, and dropping position can be accurately controlled, and minute samples (up to 30 pL/shot) can be carried out by direct administration. In addition, CPPs, which have excellent cell membrane penetration functions, can deliver high-molecular-weight drugs and nanoparticles that are difficult to penetrate through the cell membrane. By using the inkjet system, the CPPs with biofunctional cargo, including peptides, proteins such as antibodies, and exosomes, could be accurately delivered to cells, and efficient cytosolic transduction was confirmed.
Collapse
Affiliation(s)
- Mika Omura
- Department
of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Kenta Morimoto
- Department
of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Yurina Araki
- Department
of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Department
of Biological Chemistry, School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Hisaaki Hirose
- Institute
for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yoshimasa Kawaguchi
- Institute
for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yukiya Kitayama
- Department
of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Yuto Goto
- Department
of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Atsushi Harada
- Department
of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Ikuo Fujii
- Department
of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Department
of Biological Chemistry, School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| | - Tomoka Takatani-Nakase
- Department
of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, 11-68, Koshien Kyuban-cho, Nishinomiya 663-8179, Hyogo, Japan
- Institute
for Bioscience, Mukogawa Women’s
University, 11-68, Koshien
Kyuban-cho, Nishinomiya 663-8179, Hyogo, Japan
| | - Shiroh Futaki
- Institute
for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Ikuhiko Nakase
- Department
of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
- Department
of Biological Chemistry, School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai 599-8531, Osaka, Japan
| |
Collapse
|
7
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
8
|
Garner AL, Neculaes B, Dylov DV. Infrared Laser-Based Single Cell Permeabilization by Plasma Membrane Temperature Gradients. MEMBRANES 2022; 12:membranes12060574. [PMID: 35736281 PMCID: PMC9227360 DOI: 10.3390/membranes12060574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/25/2022] [Accepted: 05/29/2022] [Indexed: 01/27/2023]
Abstract
Single cell microinjection provides precise tuning of the volume and timing of delivery into the treated cells; however, it also introduces workflow complexity that requires highly skilled operators and specialized equipment. Laser-based microinjection provides an alternative method for targeting a single cell using a common laser and a workflow that may be readily standardized. This paper presents experiments using a 1550 nm, 100 fs pulse duration laser with a repetition rate of 20 ns for laser-based microinjection and calculations of the hypothesized physical mechanism responsible for the experimentally observed permeabilization. Chinese Hamster Ovarian (CHO) cells exposed to this laser underwent propidium iodide uptake, demonstrating the potential for selective cell permeabilization. The agreement between the experimental conditions and the electropermeabilization threshold based on estimated changes in the transmembrane potential induced by a laser-induced plasma membrane temperature gradient, even without accounting for enhancement due to traditional electroporation, strengthens the hypothesis of this mechanism for the experimental observations. Compared to standard 800 nm lasers, 1550 nm fs lasers may ultimately provide a lower cost microinjection method that readily interfaces with a microscope and is agnostic to operator skill, while inducing fewer deleterious effects (e.g., temperature rise, shockwaves, and cavitation bubbles).
Collapse
Affiliation(s)
- Allen L. Garner
- School of Nuclear Engineering, Purdue University, West Lafayette, IN 47906, USA
- Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (A.L.G.); (B.N.)
| | - Bogdan Neculaes
- GE Research, Niskayuna, NY 12309, USA;
- Correspondence: (A.L.G.); (B.N.)
| | | |
Collapse
|
9
|
An Overview of Cell Membrane Perforation and Resealing Mechanisms for Localized Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14040886. [PMID: 35456718 PMCID: PMC9031838 DOI: 10.3390/pharmaceutics14040886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/09/2022] [Accepted: 04/14/2022] [Indexed: 01/04/2023] Open
Abstract
Localized and reversible plasma membrane disruption is a promising technique employed for the targeted deposition of exogenous therapeutic compounds for the treatment of disease. Indeed, the plasma membrane represents a significant barrier to successful delivery, and various physical methods using light, sound, and electrical energy have been developed to generate cell membrane perforations to circumvent this issue. To restore homeostasis and preserve viability, localized cellular repair mechanisms are subsequently triggered to initiate a rapid restoration of plasma membrane integrity. Here, we summarize the known emergency membrane repair responses, detailing the salient membrane sealing proteins as well as the underlying cytoskeletal remodeling that follows the physical induction of a localized plasma membrane pore, and we present an overview of potential modulation strategies that may improve targeted drug delivery approaches.
Collapse
|
10
|
Qian C, Yu X, Tong M, Zhuang S, Lin W. Visual-Guided Solutions in Automated Zebrafish Larva Heart Micro-Injection. IEEE Robot Autom Lett 2022. [DOI: 10.1109/lra.2021.3140059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Cheng Qian
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| | - Xinghu Yu
- Ningbo Institute of Intelligent Equipment Technology Co. Ltd., Ningbo, China
| | - Mingsi Tong
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| | - Songlin Zhuang
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada
| | - Weiyang Lin
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
11
|
Xu Z, Wang Q, Zhong H, Jiang Y, Shi X, Yuan B, Yu N, Zhang S, Yuan X, Guo S, Yang Y. Carrier strategies boost the application of CRISPR/Cas system in gene therapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210081. [PMID: 37323878 PMCID: PMC10190933 DOI: 10.1002/exp.20210081] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023]
Abstract
Emerging clustered regularly interspaced short palindromic repeat/associated protein (CRISPR/Cas) genome editing technology shows great potential in gene therapy. However, proteins and nucleic acids suffer from enzymatic degradation in the physiological environment and low permeability into cells. Exploiting carriers to protect the CRISPR system from degradation, enhance its targeting of specific tissues and cells, and reduce its immunogenicity is essential to stimulate its clinical applications. Here, the authors review the state-of-the-art CRISPR delivery systems and their applications, and describe strategies to improve the safety and efficacy of CRISPR mediated genome editing, categorized by three types of cargo formats, that is, Cas: single-guide RNA ribonucleoprotein, Cas mRNA and single-guide RNA, and Cas plasmid expressing CRISPR/Cas systems. The authors hope this review will help develop safe and efficient nanomaterial-based carriers for CRISPR tools.
Collapse
Affiliation(s)
- Zunkai Xu
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| | - Haiping Zhong
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yaoyao Jiang
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Xiaoguang Shi
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Bo Yuan
- School of MedicineNankai UniversityTianjinChina
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
| | - Na Yu
- Translational Medicine CenterKey Laboratory of Molecular Target & Clinical PharmacologySchool of Pharmaceutical Sciences and The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of EducationDalian Minzu UniversityDalianChina
| | - Xiaoyong Yuan
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
- Clinical College of OphthalmologyTianjin Medical UniversityTianjinChina
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| |
Collapse
|
12
|
Chen S, Jiao Y, Pan F, Guan Z, Cheng SH, Sun D. Knock-in of a Large Reporter Gene via the High-Throughput Microinjection of the CRISPR/Cas9 System. IEEE Trans Biomed Eng 2022; 69:2524-2532. [PMID: 35133958 DOI: 10.1109/tbme.2022.3149530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The non-viral delivery of the prokaryotic clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) nuclease system provides promising solutions for gene therapy. However, traditional chemical and physical delivery approaches for gene knock-in are confronted by significant challenges to overcome the draw-backs of low efficiency and high toxicity. An alternative method for directly delivering CRISPR components into single cells is microinjection. Here, we present the high-throughput robotic microinjection of CRISPR machinery plasmids to produce gene insertions. We demonstrate that the microinjection of CRISPR/Cas9 with an enhanced green fluorescent protein (eGFP) donor template into single HepG2 cells can achieve re-porter gene knock-in targeting the adeno-associated virus site 1 locus. Homology-directed repair-mediated knock-in can be ob-served with an efficiency of 41%. Assessment via T7E1 assay indicates that the eGFP knock-in cells exhibit no detectable changes at potential off-target sites. A case study of injecting the eGFP knock-in cells into zebrafish (Danio rerio) embryos to form an in vivo tumor model is conducted. Results demonstrate the efficiency of combining microinjection with the CRISPR/Cas9 system in achieving gene knock-in.
Collapse
|
13
|
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: 4.7] [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
|
14
|
Muthaiyan Shanmugam M, Manoj H. Microinjection for Single-Cell Analysis and Therapy. HANDBOOK OF SINGLE-CELL TECHNOLOGIES 2022:81-107. [DOI: 10.1007/978-981-10-8953-4_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Qian C, Tong M, Yu X, Zhuang S. CNN-based visual processing approach for biological sample microinjection systems. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2021.06.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
16
|
Sui B, Wang M, Cheng C, Zhang Q, Zhang J, Fan D, Xu P. Nanogel-facilitated Protein Intracellular Specific Degradation through Trim-Away. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2010556. [PMID: 34421476 PMCID: PMC8376022 DOI: 10.1002/adfm.202010556] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 05/10/2023]
Abstract
Recently discovered "Trim-Away" mechanism opens a new window for fast and selective degradation of endogenous proteins. However, the in vivo and clinical application of this approach is stuck by the requirement of special skills and equipment needed for the intracellular delivery of antibodies. Hereby, an antibody conjugated polymer nanogel system, Nano-ERASER, for intracellular delivery and release of antibody, and degradation of a specific endogenous protein has been developed. After being delivered into cells, the antibody is released and forms complex with its target protein, and subsequently binds to the Fc receptor of TRIM21. The resulted complex of target protein/antibody/TRIM21 is then degraded by the proteasome. The efficacy of Nano-ERASER has been validated by depleting GFP protein in a GFP expressing cell line. Furthermore, Nano-ERASER successfully degrades COPZ1, a vital protein for cancer cells, and kills those cells while sparing normal cells. Benefit from its convenience and targeted delivery merit, Nano-ERASER technique is promising in providing a reliable tool for endogenous protein function study as well as paves the way for novel antibody-based Trim-Away therapeutic modalities for cancer and other diseases.
Collapse
Affiliation(s)
- Binglin Sui
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208 (USA)
| | - Mingming Wang
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208 (USA)
| | - Chen Cheng
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208 (USA)
| | - Quanguang Zhang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, University of South Carolina
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina
| | - Peisheng Xu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208 (USA)
| |
Collapse
|
17
|
Fus-Kujawa A, Prus P, Bajdak-Rusinek K, Teper P, Gawron K, Kowalczuk A, Sieron AL. An Overview of Methods and Tools for Transfection of Eukaryotic Cells in vitro. Front Bioeng Biotechnol 2021; 9:701031. [PMID: 34354988 PMCID: PMC8330802 DOI: 10.3389/fbioe.2021.701031] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Transfection is a powerful analytical tool enabling studies of gene products and functions in eukaryotic cells. Successful delivery of genetic material into cells depends on DNA quantity and quality, incubation time and ratio of transfection reagent to DNA, the origin, type and the passage of transfected cells, and the presence or absence of serum in the cell culture. So far a number of transfection methods that use viruses, non-viral particles or physical factors as the nucleic acids carriers have been developed. Among non-viral carriers, the cationic polymers are proposed as the most attractive ones due to the possibility of their chemical structure modification, low toxicity and immunogenicity. In this review the delivery systems as well as physical, biological and chemical methods used for eukaryotic cells transfection are described and discussed.
Collapse
Affiliation(s)
- Agnieszka Fus-Kujawa
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Pawel Prus
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Students’ Scientific Society, Katowice, Poland
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Paulina Teper
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Katarzyna Gawron
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Aleksander L. Sieron
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
18
|
Sahmi-Bounsiar D, Baudoin JP, Hannat S, Decloquement P, Chabrieres E, Aherfi S, La Scola B. Generation of Infectious Mimivirus Virions Through Inoculation of Viral DNA Within Acanthamoeba castellanii Shows Involvement of Five Proteins, Essentially Uncharacterized. Front Microbiol 2021; 12:677847. [PMID: 34305841 PMCID: PMC8299487 DOI: 10.3389/fmicb.2021.677847] [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: 03/08/2021] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
One of the most curious findings associated with the discovery of Acanthamoeba polyphaga mimivirus (APMV) was the presence of many proteins and RNAs within the virion. Although some hypotheses on their role in Acanthamoeba infection have been put forward, none have been validated. In this study, we directly transfected mimivirus DNA with or without additional proteinase K treatment to extracted DNA into Acanthamoeba castellanii. In this way, it was possible to generate infectious APMV virions, but only without extra proteinase K treatment of extracted DNA. The virus genomes before and after transfection were identical. We searched for the remaining DNA-associated proteins that were digested by proteinase K and could visualize at least five putative proteins. Matrix-assisted laser desorption/ionization time-of-flight and liquid chromatography–mass spectrometry comparison with protein databases allowed the identification of four hypothetical proteins—L442, L724, L829, and R387—and putative GMC-type oxidoreductase R135. We believe that L442 plays a major role in this protein–DNA interaction. In the future, expression in vectors and then diffraction of X-rays by protein crystals could help reveal the exact structure of this protein and its precise role.
Collapse
Affiliation(s)
- Dehia Sahmi-Bounsiar
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Jean-Pierre Baudoin
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Sihem Hannat
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Philippe Decloquement
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Eric Chabrieres
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Sarah Aherfi
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Bernard La Scola
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille Université, Institut de Recherche pour le Développement (IRD), Assistance Publique- Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| |
Collapse
|
19
|
Kaladharan K, Kumar A, Gupta P, Illath K, Santra TS, Tseng FG. Microfluidic Based Physical Approaches towards Single-Cell Intracellular Delivery and Analysis. MICROMACHINES 2021; 12:631. [PMID: 34071732 PMCID: PMC8228766 DOI: 10.3390/mi12060631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022]
Abstract
The ability to deliver foreign molecules into a single living cell with high transfection efficiency and high cell viability is of great interest in cell biology for applications in therapeutic development, diagnostics, and drug delivery towards personalized medicine. Various physical delivery methods have long demonstrated the ability to deliver cargo molecules directly to the cytoplasm or nucleus and the mechanisms underlying most of the approaches have been extensively investigated. However, most of these techniques are bulk approaches that are cell-specific and have low throughput delivery. In comparison to bulk measurements, single-cell measurement technologies can provide a better understanding of the interactions among molecules, organelles, cells, and the microenvironment, which can aid in the development of therapeutics and diagnostic tools. To elucidate distinct responses during cell genetic modification, methods to achieve transfection at the single-cell level are of great interest. In recent years, single-cell technologies have become increasingly robust and accessible, although limitations exist. This review article aims to cover various microfluidic-based physical methods for single-cell intracellular delivery such as electroporation, mechanoporation, microinjection, sonoporation, optoporation, magnetoporation, and thermoporation and their analysis. The mechanisms of various physical methods, their applications, limitations, and prospects are also elaborated.
Collapse
Affiliation(s)
- Kiran Kaladharan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| | - Ashish Kumar
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| |
Collapse
|
20
|
Morshedi Rad D, Alsadat Rad M, Razavi Bazaz S, Kashaninejad N, Jin D, Ebrahimi Warkiani M. A Comprehensive Review on Intracellular Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005363. [PMID: 33594744 DOI: 10.1002/adma.202005363] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/22/2020] [Indexed: 05/22/2023]
Abstract
Intracellular delivery is considered an indispensable process for various studies, ranging from medical applications (cell-based therapy) to fundamental (genome-editing) and industrial (biomanufacture) approaches. Conventional macroscale delivery systems critically suffer from such issues as low cell viability, cytotoxicity, and inconsistent material delivery, which have opened up an interest in the development of more efficient intracellular delivery systems. In line with the advances in microfluidics and nanotechnology, intracellular delivery based on micro- and nanoengineered platforms has progressed rapidly and held great promises owing to their unique features. These approaches have been advanced to introduce a smorgasbord of diverse cargoes into various cell types with the maximum efficiency and the highest precision. This review differentiates macro-, micro-, and nanoengineered approaches for intracellular delivery. The macroengineered delivery platforms are first summarized and then each method is categorized based on whether it employs a carrier- or membrane-disruption-mediated mechanism to load cargoes inside the cells. Second, particular emphasis is placed on the micro- and nanoengineered advances in the delivery of biomolecules inside the cells. Furthermore, the applications and challenges of the established and emerging delivery approaches are summarized. The topic is concluded by evaluating the future perspective of intracellular delivery toward the micro- and nanoengineered approaches.
Collapse
Affiliation(s)
- Dorsa Morshedi Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Maryam Alsadat Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Navid Kashaninejad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Institute of Molecular Medicine, Sechenov University, Moscow, 119991, Russia
| |
Collapse
|
21
|
Shinde A, Illath K, Gupta P, Shinde P, Lim KT, Nagai M, Santra TS. A Review of Single-Cell Adhesion Force Kinetics and Applications. Cells 2021; 10:577. [PMID: 33808043 PMCID: PMC8000588 DOI: 10.3390/cells10030577] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Cells exert, sense, and respond to the different physical forces through diverse mechanisms and translating them into biochemical signals. The adhesion of cells is crucial in various developmental functions, such as to maintain tissue morphogenesis and homeostasis and activate critical signaling pathways regulating survival, migration, gene expression, and differentiation. More importantly, any mutations of adhesion receptors can lead to developmental disorders and diseases. Thus, it is essential to understand the regulation of cell adhesion during development and its contribution to various conditions with the help of quantitative methods. The techniques involved in offering different functionalities such as surface imaging to detect forces present at the cell-matrix and deliver quantitative parameters will help characterize the changes for various diseases. Here, we have briefly reviewed single-cell mechanical properties for mechanotransduction studies using standard and recently developed techniques. This is used to functionalize from the measurement of cellular deformability to the quantification of the interaction forces generated by a cell and exerted on its surroundings at single-cell with attachment and detachment events. The adhesive force measurement for single-cell microorganisms and single-molecules is emphasized as well. This focused review should be useful in laying out experiments which would bring the method to a broader range of research in the future.
Collapse
Affiliation(s)
- Ashwini Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon-Si, Gangwon-Do 24341, Korea;
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi 441-8580, Japan;
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| |
Collapse
|
22
|
|
23
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
24
|
Abstract
Fluorescence microscopy is advantageous for investigating biological processes and mechanisms in living cells. One of the most important considerations when designing an experiment is the selection of an appropriate fluorescent probe. Equally important is deciding how the probe will be attached to the protein of interest. The advantages and disadvantages of different fluorescent probe types and their respective labeling methods are discussed to provide an overview on selecting appropriate fluorophores and labeling systems for fluorescence-based assays. Protocols are outlined when appropriate.
Collapse
|
25
|
Babu Reddiar S, Al-Wassiti H, Pouton CW, Nowell CJ, Matthews MA, Rahman A, Barlow N, Norton RS. Assessing the cellular toxicity of peptide inhibitors of intracellular protein-protein interactions by microinjection. Bioorg Med Chem 2021; 29:115906. [PMID: 33310547 DOI: 10.1016/j.bmc.2020.115906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 10/22/2022]
Abstract
Inhibitors of protein-protein interactions can be developed through a number of technologies to provide leads that include cell-impermeable molecules. Redesign of these impermeable leads to provide cell-permeable derivatives can be challenging and costly. We hypothesised that intracellular toxicity of leads could be assessed by microinjection prior to investing in the redesign process. We demonstrate this approach for our development of inhibitors of the protein-protein interaction between inducible nitric-oxide synthase (iNOS) and SPRY domain-containing SOCS box proteins (SPSBs). We microinjected a lead molecule into AD-293 cells and were able to perform an intracellular toxicity assessment. We also investigated the intracellular distribution and localisation of injected inhibitor using a fluorescently-labelled analogue. Our findings show that a lead peptide inhibitor, CP2, had no toxicity even at intracellular concentrations four orders of magnitude higher than its Kd for binding to SPSB2. This early toxicity assessment justifies further development of this cell-impermeable lead to confer cell permeability. Our investigation highlights the utility of microinjection as a tool for assessing toxicity during development of drugs targeting protein-protein interactions.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Hareth Al-Wassiti
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Colin W Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Macgregor A Matthews
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Arfatur Rahman
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia
| | - Nicholas Barlow
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia.
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Vic, Australia; ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia.
| |
Collapse
|
26
|
Brooks J, Minnick G, Mukherjee P, Jaberi A, Chang L, Espinosa HD, Yang R. High Throughput and Highly Controllable Methods for In Vitro Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004917. [PMID: 33241661 PMCID: PMC8729875 DOI: 10.1002/smll.202004917] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/06/2020] [Indexed: 05/03/2023]
Abstract
In vitro and ex vivo intracellular delivery methods hold the key for releasing the full potential of tissue engineering, drug development, and many other applications. In recent years, there has been significant progress in the design and implementation of intracellular delivery systems capable of delivery at the same scale as viral transfection and bulk electroporation but offering fewer adverse outcomes. This review strives to examine a variety of methods for in vitro and ex vivo intracellular delivery such as flow-through microfluidics, engineered substrates, and automated probe-based systems from the perspective of throughput and control. Special attention is paid to a particularly promising method of electroporation using micro/nanochannel based porous substrates, which expose small patches of cell membrane to permeabilizing electric field. Porous substrate electroporation parameters discussed include system design, cells and cargos used, transfection efficiency and cell viability, and the electric field and its effects on molecular transport. The review concludes with discussion of potential new innovations which can arise from specific aspects of porous substrate-based electroporation platforms and high throughput, high control methods in general.
Collapse
Affiliation(s)
- Justin Brooks
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Grayson Minnick
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Arian Jaberi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Horacio D. Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
27
|
He W, Xing X, Wang X, Wu D, Wu W, Guo J, Mitragotri S. Nanocarrier‐Mediated Cytosolic Delivery of Biopharmaceuticals. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910566] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/16/2020] [Indexed: 01/04/2025]
Abstract
AbstractBiopharmaceuticals have emerged to play a vital role in disease treatment and have shown promise in the rapidly expanding pharmaceutical market due to their high specificity and potency. However, the delivery of these biologics is hindered by various physiological barriers, owing primarily to the poor cell membrane permeability, low stability, and increased size of biologic agents. Since many biological drugs are intended to function by interacting with intracellular targets, their delivery to intracellular targets is of high relevance. In this review, the authors summarize and discuss the use of nanocarriers for intracellular delivery of biopharmaceuticals via endosomal escape and, especially, the routes of direct cytosolic delivery by means including the caveolae‐mediated pathway, contact release, intermembrane transfer, membrane fusion, direct translocation, and membrane disruption. Strategies with high potential for translation are highlighted. Finally, the authors conclude with the clinical translation of promising carriers and future perspectives.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xuyang Xing
- Department of Pharmaceutics School of Pharmacy China Pharmaceutical University Nanjing 210009 China
| | - Xiaoling Wang
- School of Biomass Science and Engineering Sichuan University Chengdu 610065 China
| | - Debra Wu
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of Ministry of Education of China School of Pharmacy Fudan University Shanghai 201203 China
| | - Junling Guo
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| |
Collapse
|
28
|
Neural networks-based model predictive control for precision motion tracking of a micropositioning system. INTERNATIONAL JOURNAL OF INTELLIGENT ROBOTICS AND APPLICATIONS 2020. [DOI: 10.1007/s41315-020-00134-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Pan F, Chen S, Jiao Y, Guan Z, Shakoor A, Sun D. Automated High-Productivity Microinjection System for Adherent Cells. IEEE Robot Autom Lett 2020. [DOI: 10.1109/lra.2020.2965870] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Ki B, Choi K, Kim K, Oh J. Electrochemical local etching of silicon in etchant vapor. NANOSCALE 2020; 12:6411-6419. [PMID: 32141459 DOI: 10.1039/c9nr10420h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Direct machining and imprinting of Si are beneficial for simplifying the fabrication of microelectromechanical systems, nanoelectromechanical systems, optical devices, and fin field-effect transistors, and for reducing process costs. Electrochemical micromachining has been introduced for highly doped Si, but complex structures cannot be imprinted directly. With chemical imprinting, complex nano/micropatterns can be imprinted even on low-doped Si, but the physical contact can damage the templates. In this study, we demonstrated an electrochemical local etching (ELE) method for fabricating nano/micrometer structures on semiconductors in a noncontact manner. Polygon tips were prepared as templates on highly doped n-type Si via etching in KOH. A constant space is maintained between the template and the target Si using a gap layer to prevent damage and contamination. In the etchant vapor, the voltage bias between the template and the target Si leads to condensation of the etchant. Because the etching region is localized by the condensation of the etchant, even low-doped semiconductors can be imprinted in submicrometer patterns in a single step. When the etchant condensation is suppressed, the etching area is reduced and the resolution is increased, allowing direct imprinting of the polygonal submicrometer pattern. ELE has the potential to produce complex nano/micrometer structures in a single step without photoresists and physical contact.
Collapse
Affiliation(s)
- Bugeun Ki
- School of Integrated Technology, Yonsei University, Incheon 21983, Republic of Korea. and Yonsei Institute of Convergence Technology, Incheon 21983, Republic of Korea
| | - Keorock Choi
- School of Integrated Technology, Yonsei University, Incheon 21983, Republic of Korea. and Yonsei Institute of Convergence Technology, Incheon 21983, Republic of Korea
| | - Kyunghwan Kim
- School of Integrated Technology, Yonsei University, Incheon 21983, Republic of Korea. and Yonsei Institute of Convergence Technology, Incheon 21983, Republic of Korea
| | - Jungwoo Oh
- School of Integrated Technology, Yonsei University, Incheon 21983, Republic of Korea. and Yonsei Institute of Convergence Technology, Incheon 21983, Republic of Korea
| |
Collapse
|
31
|
Kopytynski M, Chen S, Legg S, Minter R, Chen R. A Versatile Polymer‐Based Platform for Intracellular Delivery of Macromolecules. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Michal Kopytynski
- Department of Chemical EngineeringImperial College London South Kensington Campus London SW7 2AZ UK
| | - Siyuan Chen
- Department of Chemical EngineeringImperial College London South Kensington Campus London SW7 2AZ UK
| | - Sandrine Legg
- Department of Antibody Discovery and Protein EngineeringAstraZeneca Milstein Building, Granta Park Cambridge CB21 6GH UK
| | - Ralph Minter
- Department of Antibody Discovery and Protein EngineeringAstraZeneca Milstein Building, Granta Park Cambridge CB21 6GH UK
| | - Rongjun Chen
- Department of Chemical EngineeringImperial College London South Kensington Campus London SW7 2AZ UK
| |
Collapse
|
32
|
Chang L, Wang YC, Ershad F, Yang R, Yu C, Fan Y. Wearable Devices for Single-Cell Sensing and Transfection. Trends Biotechnol 2019; 37:1175-1188. [DOI: 10.1016/j.tibtech.2019.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/31/2019] [Accepted: 04/02/2019] [Indexed: 02/01/2023]
|
33
|
Shull G, Haffner C, Huttner WB, Kodandaramaiah SB, Taverna E. Robotic platform for microinjection into single cells in brain tissue. EMBO Rep 2019; 20:e47880. [PMID: 31469223 PMCID: PMC6776899 DOI: 10.15252/embr.201947880] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/23/2019] [Accepted: 08/07/2019] [Indexed: 01/02/2023] Open
Abstract
Microinjection into single cells in brain tissue is a powerful technique to study and manipulate neural stem cells. However, such microinjection requires expertise and is a low-throughput process. We developed the "Autoinjector", a robot that utilizes images from a microscope to guide a microinjection needle into tissue to deliver femtoliter volumes of liquids into single cells. The Autoinjector enables microinjection of hundreds of cells within a single organotypic slice, resulting in an overall yield that is an order of magnitude greater than manual microinjection. The Autoinjector successfully targets both apical progenitors (APs) and newborn neurons in the embryonic mouse and human fetal telencephalon. We used the Autoinjector to systematically study gap-junctional communication between neural progenitors in the embryonic mouse telencephalon and found that apical contact is a characteristic feature of the cells that are part of a gap junction-coupled cluster. The throughput and versatility of the Autoinjector will render microinjection an accessible high-performance single-cell manipulation technique and will provide a powerful new platform for performing single-cell analyses in tissue for bioengineering and biophysics applications.
Collapse
Affiliation(s)
- Gabriella Shull
- Department of Biomedical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
- Department of Biomedical EngineeringDuke UniversityDurhamNCUSA
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Suhasa B Kodandaramaiah
- Department of Biomedical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
- Department of Mechanical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
| | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| |
Collapse
|
34
|
Singh K, Ejaz W, Dutta K, Thayumanavan S. Antibody Delivery for Intracellular Targets: Emergent Therapeutic Potential. Bioconjug Chem 2019; 30:1028-1041. [PMID: 30830750 PMCID: PMC6470022 DOI: 10.1021/acs.bioconjchem.9b00025] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins have sparked fast growing interest as biological therapeutic agents for several diseases. Antibodies, in particular, carry an enormous potential as drugs owing to their remarkable target specificity and low immunogenicity. Although the market has numerous antibodies directed toward extracellular targets, their use in targeting therapeutically important intracellular targets is limited by their inability to cross cellular membrane. Realizing the potential for antibody therapy in disease treatment, progress has been made in the development of methods to deliver antibodies intracellularly. In this review, we address various platforms for delivery of antibodies and their merits and drawbacks.
Collapse
|
35
|
Doll F, Steimbach RR, Zumbusch A. Direct Imaging of Protein‐Specific Methylation in Mammalian Cells. Chembiochem 2019; 20:1315-1325. [DOI: 10.1002/cbic.201800787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Franziska Doll
- Department of ChemistryUniversity of Konstanz Universitätsstrasse 10 78457 Konstanz Germany
- Konstanz Research School Chemical Biology Universitätsstrasse 10 78457 Konstanz Germany
| | - Raphael R. Steimbach
- Department of ChemistryUniversity of Konstanz Universitätsstrasse 10 78457 Konstanz Germany
| | - Andreas Zumbusch
- Department of ChemistryUniversity of Konstanz Universitätsstrasse 10 78457 Konstanz Germany
- Konstanz Research School Chemical Biology Universitätsstrasse 10 78457 Konstanz Germany
| |
Collapse
|
36
|
Loureiro A, da Silva GJ. CRISPR-Cas: Converting A Bacterial Defence Mechanism into A State-of-the-Art Genetic Manipulation Tool. Antibiotics (Basel) 2019; 8:E18. [PMID: 30823430 PMCID: PMC6466564 DOI: 10.3390/antibiotics8010018] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Bacteriophages are pervasive viruses that infect bacteria, relying on their genetic machinery to replicate. In order to protect themselves from this kind of invader, bacteria developed an ingenious adaptive defence system, clustered regularly interspaced short palindromic repeats (CRISPR). Researchers soon realised that a specific type of CRISPR system, CRISPR-Cas9, could be modified into a simple and efficient genetic engineering technology, with several improvements over currently used systems. This discovery set in motion a revolution in genetics, with new and improved CRISPR systems being used in plenty of in vitro and in vivo experiments in recent years. This review illustrates the mechanisms behind CRISPR-Cas systems as a means of bacterial immunity against phage invasion and how these systems were engineered to originate new genetic manipulation tools. Newfound CRISPR-Cas technologies and the up-and-coming applications of these systems on healthcare and other fields of science are also discussed.
Collapse
Affiliation(s)
- Alexandre Loureiro
- Laboratory of Microbiology, Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - Gabriela Jorge da Silva
- Laboratory of Microbiology, Faculty of Pharmacy, University of Coimbra, Health Sciences Campus, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
- Center for Neurosciences Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
37
|
Moore CL, Taylor EM, Ball KK, Bernock LJ, Griffin RJ, Jung S, Shoeib A, Borrelli MJ. Quantitative microinjection using fluorescence calibration of streaming microdroplets on a superhydrophobic surface. Exp Cell Res 2018; 370:426-433. [PMID: 29981341 DOI: 10.1016/j.yexcr.2018.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
Abstract
A simple and reproducible procedure was developed to measure the volume of liquid microinjected into cells. A calibration curve of droplet fluorescence intensity versus volume was constructed by injecting a fluorescent dextran solution through a 125-150 µm diameter micropipette into an oil-filled culture dish to create a spray of varied-sized droplets. The droplets retained a spherical shape because they were in an oil medium and they settled onto a glass surface coated with a superhydrophobic surface. Fluorescent micrographs of the droplets were obtained and analyzed with Image-J software to quantify the fluorescence intensity and radius of each spherical droplet to produce the calibration curve. Subsequently, Dut-145 human prostate carcinoma cells were microinjected with the same fluorescent dextran solution and fluorescent micrographs of the cells were obtained using the identical exposure conditions used to photograph the droplets. The measured fluorescence intensity of the microinjected cells was entered into the formula for the regression line that was fit to the calibration curve allowing determination of the volume of solution injected into each cell. Thus, a mixture consisting of known concentrations of a test material of test material (macromolecules, drugs, etc.) and a fluorescent dextran, volumetric, tracer can be used to quantify the relationship between the amount of a microinjected material and subsequent effects on cells.
Collapse
Affiliation(s)
- Christopher L Moore
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Erin M Taylor
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Kelly K Ball
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Laura J Bernock
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Robert J Griffin
- Departments of Radiation Oncology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Seunghyun Jung
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Amal Shoeib
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| | - Michael J Borrelli
- Departments of Radiology, Center for Advanced Surface Engineering, Arkansas Nanomedicine Center, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA.
| |
Collapse
|
38
|
Zhang N, Chin JS, Chew SY. Localised non-viral delivery of nucleic acids for nerve regeneration in injured nervous systems. Exp Neurol 2018; 319:112820. [PMID: 30195695 DOI: 10.1016/j.expneurol.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
Axons damaged by traumatic injuries are often unable to spontaneously regenerate in the adult central nervous system (CNS). Although the peripheral nervous system (PNS) has some regenerative capacity, its ability to regrow remains limited across large lesion gaps due to scar tissue formation. Nucleic acid therapy holds the potential of improving regeneration by enhancing the intrinsic growth ability of neurons and overcoming the inhibitory environment that prevents neurite outgrowth. Nucleic acids modulate gene expression by over-expression of neuronal growth factor or silencing growth-inhibitory molecules. Although in vitro outcomes appear promising, the lack of efficient non-viral nucleic acid delivery methods to the nervous system has limited the application of nucleic acid therapeutics to patients. Here, we review the recent development of efficient non-viral nucleic acid delivery platforms, as applied to the nervous system, including the transfection vectors and carriers used, as well as matrices and scaffolds that are currently used. Additionally, we will discuss possible improvements for localised nucleic acid delivery.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; NTU Institute of Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 639798, Singapore
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore.
| |
Collapse
|
39
|
Wang X, Zhao Q, Wang L, Liu J, Pu H, Xie S, Ru C, Sun Y. Effect of Cell Inner Pressure on Deposition Volume in Microinjection. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:10287-10292. [PMID: 30095920 DOI: 10.1021/acs.langmuir.8b02102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Microinjection is a widely used technique for introducing exogenous materials into cells. Many applications of microinjection, such as gene editing and drug testing, rely on the accurate control of the deposition volume. However, the deposition volume in microinjection is presently calibrated in an open medium without considering the cell inner pressure effect, which we experimentally show in this paper that it can induce an error as large as 30% between the actual deposition volume and the set volume. In this work, the relationship between the cell inner pressure and the deposition volume was analytically modeled and experimentally validated. On the basis of the developed model, the cell inner pressure of a given cell type can be well estimated from the injection pressure and the resulting deposition volume. The quantitated cell inner pressure is then used to reduce the error between the set volume and the actual deposition volume. Experiments conducted on human bladder cancer cells (T24 and RT4) showed that T24 cells have a higher inner pressure than RT4 cells (405 ± 45 Pa vs 341 ± 34 Pa), and after compensating for the cell inner pressure, the error between the intended set volume and the actual deposition volume into a cell became less than 3%.
Collapse
Affiliation(s)
- Xian Wang
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto M5S 3G8 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5S 3G9 , Canada
| | - Qili Zhao
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto M5S 3G8 , Canada
| | - Li Wang
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto M5S 3G8 , Canada
| | - Jun Liu
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto M5S 3G8 , Canada
| | - Huayan Pu
- School of Mechatronic Engineering and Automation , Shanghai University , Shanghai 200072 , China
| | - Shaorong Xie
- School of Mechatronic Engineering and Automation , Shanghai University , Shanghai 200072 , China
| | - Changhai Ru
- Research Center of Robotics and Micro System & Collaborative Innovation Center of Suzhou Nano Science and Technology , Soochow University , Suzhou , Jiangsu 215021 , China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto M5S 3G8 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto M5S 3G9 , Canada
- Department of Electrical and Computer Engineering , University of Toronto , Toronto M5S 3G4 , Canada
| |
Collapse
|
40
|
Yang# Y, Yu# J, Esfahani AM, Seiffert-Sinha K, Xi N, Lee I, Sinha AA, Chen L, Sun Z, Yang R, Dong L. Single-cell membrane drug delivery using porous pen nanodeposition. NANOSCALE 2018; 10:12704-12712. [PMID: 29946596 PMCID: PMC6528655 DOI: 10.1039/c8nr02600a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Delivering molecules onto the plasma membrane of single cells is still a challenging task in profiling cell signaling pathways with single cell resolution. We demonstrated that a large quantity of molecules could be targeted and released onto the membrane of individual cells to trigger signaling responses. This is achieved by a porous pen nanodeposition (PPN) method, in which a multilayer porous structure, serving as a reservoir for a large amount of molecules, is formed on an atomic force microscope (AFM) tip using layer-by-layer assembly and post processing. To demonstrate its capability for single cell membrane drug delivery, PPN was employed to induce a calcium flux triggered by the binding of released antibodies to membrane antigens in an autoimmune skin disease model. This calcium signal propagates from the target cell to its neighbors in a matter of seconds, proving the theory of intercellular communication through cell-cell junctions. Collectively, these results demonstrated the effectiveness of PPN in membrane drug delivery for single cells; to the best of our knowledge, this is the first technique that can perform the targeted transport and delivery in single cell resolution, paving the way for probing complex signaling interactions in multicellular settings.
Collapse
Affiliation(s)
- Yongliang Yang#
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA,
| | - Jing Yu#
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska -Lincoln, Lincoln, NE 68588, USA
| | | | - Ning Xi
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Ilsoon Lee
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, USA
| | - Animesh A. Sinha
- Department of Dermatology, University at Buffalo, Buffalo, New York 14203, USA
| | - Liangliang Chen
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Zhiyong Sun
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska -Lincoln, Lincoln, NE 68588, USA,
| | - Lixin Dong
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, USA,
| |
Collapse
|
41
|
|
42
|
Cleves PA, Strader ME, Bay LK, Pringle JR, Matz MV. CRISPR/Cas9-mediated genome editing in a reef-building coral. Proc Natl Acad Sci U S A 2018; 115:5235-5240. [PMID: 29695630 PMCID: PMC5960312 DOI: 10.1073/pnas.1722151115] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Reef-building corals are critically important species that are threatened by anthropogenic stresses including climate change. In attempts to understand corals' responses to stress and other aspects of their biology, numerous genomic and transcriptomic studies have been performed, generating a variety of hypotheses about the roles of particular genes and molecular pathways. However, it has not generally been possible to test these hypotheses rigorously because of the lack of genetic tools for corals. Here, we demonstrate efficient genome editing using the CRISPR/Cas9 system in the coral Acropora millepora We targeted the genes encoding fibroblast growth factor 1a (FGF1a), green fluorescent protein (GFP), and red fluorescent protein (RFP). After microinjecting CRISPR/Cas9 ribonucleoprotein complexes into fertilized eggs, we detected induced mutations in the targeted genes using changes in restriction-fragment length, Sanger sequencing, and high-throughput Illumina sequencing. We observed mutations in ∼50% of individuals screened, and the proportions of wild-type and various mutant gene copies in these individuals indicated that mutation induction continued for at least several cell cycles after injection. Although multiple paralogous genes encoding green fluorescent proteins are present in A. millepora, appropriate design of the guide RNA allowed us to induce mutations simultaneously in more than one paralog. Because A. millepora larvae can be induced to settle and begin colony formation in the laboratory, CRISPR/Cas9-based gene editing should allow rigorous tests of gene function in both larval and adult corals.
Collapse
Affiliation(s)
- Phillip A Cleves
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Marie E Strader
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712
| | - Line K Bay
- Australian Institute of Marine Science, Townsville, QLD 4810, Australia
| | - John R Pringle
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305;
| | - Mikhail V Matz
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712;
| |
Collapse
|
43
|
Ahmed S, Miyawaki O, Matsumura K. Enhanced Adsorption of a Protein-Nanocarrier Complex onto Cell Membranes through a High Freeze Concentration by a Polyampholyte Cryoprotectant. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:2352-2362. [PMID: 29361227 DOI: 10.1021/acs.langmuir.7b03622] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The transportation of biomolecules into cells is of great importance in tissue engineering and as stimulation for antitumor immune cells. Previous freezing strategies at ultracold temperatures (-80 °C) used for intracellular transportation exhibit certain limitations such as extended time requirements and harsh delivery system conditions. Thus, the need remains to develop simplified methods for safe nanomaterial delivery. Here, we demonstrated a unique strategy based on the ice-crystallization-induced freeze concentration for protein intracellular delivery in combination with a polyampholyte cryoprotectant. We found that upon sustained lowering of the temperature from -6 to -20 °C over a short duration, the adsorption of proteins onto the peripheral cell membrane was markedly increased through the facile ice-crystallization-induced freeze concentration. Furthermore, we proposed a freeze concentration factor (α) that depends on the freezing-point depression and is estimated from an analysis of the fraction of frozen water. Notably, the α values of the polyampholyte cryoprotectant were 8-fold higher than those of the currently used cryoprotectant dimethyl sulfoxide (DMSO) at particular temperatures of interest. Our results illustrate that the presence of a polyampholyte cryoprotectant significantly enhanced the adsorption of the protein/nanocarrier complex onto membranes compared to that obtained with DMSO because of the high freeze concentration. The present study demonstrated the direct relationship between freezing and the penetration of proteins across the periphery of the cell membrane by means of increased concentration during freezing. These results may be useful in providing a guideline for the intracellular delivery of biomacromolecules using ice-crystallization-induced continuous freezing combined with polyampholyte cryoprotectants.
Collapse
Affiliation(s)
- Sana Ahmed
- School of Materials Science, Japan Advanced Institute of Science and Technology , Nomi, Ishikawa 923-1292, Japan
| | - Osato Miyawaki
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology , 4-5-7 Konan, Minato-ku, Tokyo 108-8477, Japan
| | - Kazuaki Matsumura
- School of Materials Science, Japan Advanced Institute of Science and Technology , Nomi, Ishikawa 923-1292, Japan
| |
Collapse
|
44
|
Probing and manipulating intracellular membrane traffic by microinjection of artificial vesicles. Proc Natl Acad Sci U S A 2017; 114:E9883-E9892. [PMID: 29087339 PMCID: PMC5699080 DOI: 10.1073/pnas.1713524114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
There is still a large gap in our understanding between the functional complexity of cells and the reconstruction of partial cellular functions in vitro from purified or engineered parts. Here we have introduced artificial vesicles of defined composition into living cells to probe the capacity of the cellular cytoplasm in dealing with foreign material and to develop tools for the directed manipulation of cellular functions. Our data show that protein-free liposomes, after variable delay times, are captured by the Golgi apparatus that is reached either by random diffusion or, in the case of large unilamellar vesicles, by microtubule-dependent transport via a dynactin/dynein motor complex. However, insertion of early endosomal SNARE proteins suffices to convert liposomes into trafficking vesicles that dock and fuse with early endosomes, thus overriding the default pathway to the Golgi. Moreover, such liposomes can be directed to mitochondria expressing simple artificial affinity tags, which can also be employed to divert endogenous trafficking vesicles. In addition, fusion or subsequent acidification of liposomes can be monitored by incorporation of appropriate chemical sensors. This approach provides an opportunity for probing and manipulating cellular functions that cannot be addressed by conventional genetic approaches. We conclude that the cellular cytoplasm has a remarkable capacity for self-organization and that introduction of such macromolecular complexes may advance nanoengineering of eukaryotic cells.
Collapse
|
45
|
Tiefenboeck P, Kim JA, Trunk F, Eicher T, Russo E, Teijeira A, Halin C, Leroux JC. Microinjection for the ex Vivo Modification of Cells with Artificial Organelles. ACS NANO 2017; 11:7758-7769. [PMID: 28777538 DOI: 10.1021/acsnano.7b01404] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Microinjection is extensively used across fields to deliver material intracellularly. Here we address the fundamental aspects of introducing exogenous organelles into cells to endow them with artificial functions. Nanocarriers encapsulating biologically active cargo or extreme intraluminal pH were injected directly into the cytosol of cells, where they bypassed subcellular processing pathways and remained intact for several days. Nanocarriers' size was found to dictate their intracellular distribution pattern upon injection, with larger vesicles adopting polarized agglomerated distributions and smaller colloids spreading evenly in the cytosol. This in turn determined the symmetry or asymmetry of their dilution following cell division, ultimately affecting the intracellular dose at a cell population level. As an example of microinjection's applicability, a cell type relevant for cell-based therapies (dendritic cells) was injected with vesicles, and its migratory properties were studied in a co-culture system mimicking lymphatic capillaries.
Collapse
Affiliation(s)
- Peter Tiefenboeck
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Jong Ah Kim
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Ferdinand Trunk
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Tamara Eicher
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Erica Russo
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Alvaro Teijeira
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich , 8093 Zürich, Switzerland
| |
Collapse
|
46
|
Acoustic-transfection for genomic manipulation of single-cells using high frequency ultrasound. Sci Rep 2017; 7:5275. [PMID: 28706248 PMCID: PMC5509725 DOI: 10.1038/s41598-017-05722-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/16/2017] [Indexed: 01/21/2023] Open
Abstract
Efficient intracellular delivery of biologically active macromolecules has been a challenging but important process for manipulating live cells for research and therapeutic purposes. There have been limited transfection techniques that can deliver multiple types of active molecules simultaneously into single-cells as well as different types of molecules into physically connected individual neighboring cells separately with high precision and low cytotoxicity. Here, a high frequency ultrasound-based remote intracellular delivery technique capable of delivery of multiple DNA plasmids, messenger RNAs, and recombinant proteins is developed to allow high spatiotemporal visualization and analysis of gene and protein expressions as well as single-cell gene editing using clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein-9 nuclease (Cas9), a method called acoustic-transfection. Acoustic-transfection has advantages over typical sonoporation because acoustic-transfection utilizing ultra-high frequency ultrasound over 150 MHz can directly deliver gene and proteins into cytoplasm without microbubbles, which enables controlled and local intracellular delivery to acoustic-transfection technique. Acoustic-transfection was further demonstrated to deliver CRISPR-Cas9 systems to successfully modify and reprogram the genome of single live cells, providing the evidence of the acoustic-transfection technique for precise genome editing using CRISPR-Cas9.
Collapse
|
47
|
Chan FHL, Yang R, Lai KWC. Development of the Electric Equivalent Model for the Cytoplasmic Microinjection of Small Adherent Cells. MICROMACHINES 2017; 8:mi8070216. [PMID: 30400407 PMCID: PMC6189767 DOI: 10.3390/mi8070216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/25/2017] [Accepted: 07/04/2017] [Indexed: 11/16/2022]
Abstract
A novel approach utilizing current feedback for the cytoplasmic microinjection of biological cells is proposed. In order to realize the cytoplasmic microinjection on small adherent cells (diameter < 30 μm and thickness < 10 μm), an electrical model is built and analyzed according to the electrochemical properties of target cells. In this study, we have verified the effectiveness of the current measurement for monitoring the injection process and the study of ion channel activities for verifying the cell viability of the cells after the microinjection.
Collapse
Affiliation(s)
- Florence Hiu Ling Chan
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
- Centre for Robotics and Automation, City University of Hong Kong, Hong Kong, China.
| | - Runhuai Yang
- Department of Biomedical Engineering, Anhui Medical University, Hefei 230032, China.
| | - King Wai Chiu Lai
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, China.
- Centre for Robotics and Automation, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
48
|
Norregaard K, Metzler R, Ritter CM, Berg-Sørensen K, Oddershede LB. Manipulation and Motion of Organelles and Single Molecules in Living Cells. Chem Rev 2017; 117:4342-4375. [PMID: 28156096 DOI: 10.1021/acs.chemrev.6b00638] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The biomolecule is among the most important building blocks of biological systems, and a full understanding of its function forms the scaffold for describing the mechanisms of higher order structures as organelles and cells. Force is a fundamental regulatory mechanism of biomolecular interactions driving many cellular processes. The forces on a molecular scale are exactly in the range that can be manipulated and probed with single molecule force spectroscopy. The natural environment of a biomolecule is inside a living cell, hence, this is the most relevant environment for probing their function. In vivo studies are, however, challenged by the complexity of the cell. In this review, we start with presenting relevant theoretical tools for analyzing single molecule data obtained in intracellular environments followed by a description of state-of-the art visualization techniques. The most commonly used force spectroscopy techniques, namely optical tweezers, magnetic tweezers, and atomic force microscopy, are described in detail, and their strength and limitations related to in vivo experiments are discussed. Finally, recent exciting discoveries within the field of in vivo manipulation and dynamics of single molecule and organelles are reviewed.
Collapse
Affiliation(s)
- Kamilla Norregaard
- Cluster for Molecular Imaging, Department of Biomedical Science and Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen , 2200 Copenhagen, Denmark
| | - Ralf Metzler
- Institute for Physics & Astronomy, University of Potsdam , 14476 Potsdam-Golm, Germany
| | - Christine M Ritter
- Niels Bohr Institute, University of Copenhagen , 2100 Copenhagen, Denmark
| | | | - Lene B Oddershede
- Niels Bohr Institute, University of Copenhagen , 2100 Copenhagen, Denmark
| |
Collapse
|
49
|
Munsell EV, Ross NL, Sullivan MO. Journey to the Center of the Cell: Current Nanocarrier Design Strategies Targeting Biopharmaceuticals to the Cytoplasm and Nucleus. Curr Pharm Des 2016; 22:1227-44. [PMID: 26675220 DOI: 10.2174/1381612822666151216151420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/15/2015] [Indexed: 01/06/2023]
Abstract
New biopharmaceutical molecules, potentially able to provide more personalized and effective treatments, are being identified through the advent of advanced synthetic biology strategies, sophisticated chemical synthesis approaches, and new analytical methods to assess biological potency. However, translation of many of these structures has been significantly limited due to the need for more efficient strategies to deliver macromolecular therapeutics to desirable intracellular sites of action. Engineered nanocarriers that encapsulate peptides, proteins, or nucleic acids are generally internalized into target cells via one of several endocytic pathways. These nanostructures, entrapped within endosomes, must navigate the intracellular milieu to orchestrate delivery to the intended destination, typically the cytoplasm or nucleus. For therapeutics active in the cytoplasm, endosomal escape continues to represent a limiting step to effective treatment, since a majority of nanocarriers trapped within endosomes are ultimately marked for enzymatic degradation in lysosomes. Therapeutics active in the nucleus have the added challenges of reaching and penetrating the nuclear envelope, and nuclear delivery remains a preeminent challenge preventing clinical translation of gene therapy applications. Herein, we review cutting-edge peptide- and polymer-based design strategies with the potential to enable significant improvements in biopharmaceutical efficacy through improved intracellular targeting. These strategies often mimic the activities of pathogens, which have developed innate and highly effective mechanisms to penetrate plasma membranes and enter the nucleus of host cells. Understanding these mechanisms has enabled advances in synthetic peptide and polymer design that may ultimately improve intracellular trafficking and bioavailability, leading to increased access to new classes of biotherapeutics.
Collapse
Affiliation(s)
| | | | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, Delaware.
| |
Collapse
|
50
|
Abstract
If the isolation, production, and clinical use of insulin marked the inception of the age of biologics as therapeutics, the convergence of molecular biology and combinatorial engineering techniques marked its coming of age. The first wave of recombinant protein-based drugs in the 1980s demonstrated emphatically that proteins could be engineered, formulated, and employed for clinical advantage. Yet despite the successes of protein-based drugs such as antibodies, enzymes, and cytokines, the druggable target space for biologics is currently restricted to targets outside the cell. Insofar as estimates place the number of proteins either secreted or with extracellular domains in the range of 8000 to 9000, this represents only one-third of the proteome and circumscribes the pathways that can be targeted for therapeutic intervention. Clearly, a major objective for this field to reach maturity is to access, interrogate, and modulate the majority of proteins found inside the cell. However, owing to the large size, complex architecture, and general cellular impermeability of existing protein-based drugs, this poses a daunting challenge. In recent years, though, advances on the two related fronts of protein engineering and drug delivery are beginning to bring this goal within reach. First, prompted by the restrictions that limit the applicability of antibodies, intense efforts have been applied to identifying and engineering smaller alternative protein scaffolds for the modulation of intracellular targets. In parallel, innovative solutions for delivering proteins to the intracellular space while maintaining their stability and functional activity have begun to yield successes. This review provides an overview of bioactive intrabodies and alternative protein scaffolds amenable to engineering for intracellular targeting and also outlines advances in protein engineering and formulation for delivery of functional proteins to the interior of the cell to achieve therapeutic action.
Collapse
Affiliation(s)
- Shane Miersch
- Banting and Best Department of Medical Research, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|