1
|
Emon S, Sakib S, Bardhan N, Saha S, Asaduzzaman M, Alam MK. Molecular dynamics of electroporation and quantitative analysis of molecular transport. J Biol Phys 2025; 51:18. [PMID: 40425985 DOI: 10.1007/s10867-025-09682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Electroporation, a widely used physical method for transiently increasing cell permeability, facilitates molecular delivery for therapeutic and research applications. While electroporation proves to be a useful process, the mechanisms of pore formation and molecular transport remain incompletely understood. This study investigates the dynamics of electropore formation in lipid bilayers using molecular dynamics (MD) simulations and subsequent molecular transport by quantitative diffusion modeling. MD simulations reveal different stages of pore formation under applied electric fields, focusing on the lipid headgroup realignment and the hydration process of the pores. An FDM (Finite Difference Method)-based transport model quantifies the transport of molecules, such as glucose, calcein and bleomycin, using pore dimensions obtained from MD simulations. The results demonstrate a size-dependent transport efficiency, with smaller molecules diffusing more rapidly than larger ones. This work underscores the synergy between atomistic simulations and macroscopic transport modeling. Also, the findings offer valuable insights for optimizing electroporation protocols and developing targeted delivery systems for drugs and genetic material.
Collapse
Affiliation(s)
- Shahariar Emon
- Department of Physics, University of Barishal, Barishal, 8200, Bangladesh
| | - Sadman Sakib
- Sher-E-Bangla Medical College, Barishal, 8200, Bangladesh
| | - Niloy Bardhan
- Department of Physics, University of Barishal, Barishal, 8200, Bangladesh
| | - Shovon Saha
- Department of Physics, University of Barishal, Barishal, 8200, Bangladesh
| | - Md Asaduzzaman
- Department of Physics, University of Barishal, Barishal, 8200, Bangladesh
| | - Md Khorshed Alam
- Department of Physics, University of Barishal, Barishal, 8200, Bangladesh.
| |
Collapse
|
2
|
Wu P, Zhang T, Zhao D, Xie Y, Huang D, Li Z, Huang Y. Microneedle-Enabled Breakthroughs in Nucleic Acid Therapeutics. Adv Healthc Mater 2025:e2501015. [PMID: 40370139 DOI: 10.1002/adhm.202501015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/30/2025] [Indexed: 05/16/2025]
Abstract
Nucleic acid therapy demonstrates great potential in cancer treatment, infectious disease prevention, and vaccine development due to its advantages, such as rapid production, long-lasting effects, and high target specificity. Although nucleic acid therapy is considered ideal for the development of novel therapeutic strategies, its clinical application still faces numerous challenges, including the lack of efficient delivery systems, insufficient drug formulation stability, and the limitations imposed by the skin barrier on drug dosage delivery. Microneedles, as an innovative transdermal drug delivery technology, can penetrate the stratum corneum and directly access the skin's microcirculation, enabling the efficient delivery of genes and drugs. This technology offers several advantages, such as ease of operation, minimally invasive and painless application, and high safety. Combining microneedle technology with nucleic acid therapy fully leverages the strengths of both approaches, significantly enhancing therapeutic efficacy and bioavailability while maximizing treatment potential. This review explores the application prospects and advantages of combining microneedle delivery systems with nucleic acid therapy.
Collapse
Affiliation(s)
- Pengfei Wu
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| | - Tian Zhang
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| | - Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou, 450000, P. R. China
| | - Yingqiu Xie
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, 010000, Kazakhstan
| | - Dong Huang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, School of Integrated Circuits, Peking University, Beijing, 100871, P. R. China
- AciMicro Medical Technology, Guangzhou, 510700, P. R. China
| | - Zhihong Li
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, School of Integrated Circuits, Peking University, Beijing, 100871, P. R. China
| | - Yuanyu Huang
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| |
Collapse
|
3
|
Arizono M, Idziak A, Nägerl UV. Live STED imaging of functional neuroanatomy. Nat Protoc 2025:10.1038/s41596-024-01132-6. [PMID: 40087378 DOI: 10.1038/s41596-024-01132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/11/2024] [Indexed: 03/17/2025]
Abstract
In the mammalian brain, a large network of excitable and modulatory cells efficiently processes, analyzes and stores vast amounts of information. The brain's anatomy influences the flow of neural information between neurons and glia, from which all thought, emotion and action arises. Consequently, one of the grand challenges in neuroscience is to uncover the finest structural details of the brain in the context of its overall architecture. Recent developments in microscopy and biosensors have enabled the investigation of brain microstructure and function with unprecedented specificity and resolution, dendritic spines being an exemplary case, which has provided deep insights into neuronal mechanisms of higher brain function, such as learning and memory. As diffraction-limited light microscopy methods cannot resolve the fine details of brain cells (the 'anatomical ground truth'), electron microscopy is used instead to contextualize functional signals. This approach can be quite unsatisfying given the fragility and dynamic nature of the structures under investigation. We have recently developed a method for combining super-resolution stimulated emission depletion microscopy with functional measurements in brain slices, offering nanoscale resolution in functioning brain structures. We describe how to concurrently perform morphological and functional imaging with a confocal STED microscope. Specifically, the procedure guides the user on how to record astrocytic Ca2+ signals at tripartite synapses, outlining a framework for analyzing structure-function relationships of brain cells at nanoscale resolution. The imaging requires 2-3 h and the image analysis between 2 h and 2 d.
Collapse
Affiliation(s)
- Misa Arizono
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France.
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan.
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Agata Idziak
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - U Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR 5297, Bordeaux, France.
- Department of Anatomy and Cell Biology, University Medical Center, Georg-August-University of Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Bosch M, Castro J, Sur M, Hayashi Y. Photomarking Relocalization Technique for Correlated Two-Photon and Electron Microscopy Imaging of Single Stimulated Synapses. Methods Mol Biol 2025; 2910:145-175. [PMID: 40220099 DOI: 10.1007/978-1-0716-4446-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Synapses learn and remember by persistent modifications of their internal structures and composition but, due to their small size, it is difficult to observe these changes at the ultrastructural level in real time. Two-photon fluorescence microscopy (2PM) allows time-course live imaging of individual synapses but lacks ultrastructural resolution. Electron microscopy (EM) allows the ultrastructural imaging of subcellular components but cannot detect fluorescence and lacks temporal resolution. Here we describe a combination of procedures designed to achieve the correlated imaging of the same individual synapse under both 2PM and EM. This technique permits the selective stimulation and live imaging of a single dendritic spine and the subsequent localization of the same spine in EM ultrathin serial sections. Landmarks created through a photomarking method based on the 2-photon-induced precipitation of an electrodense compound are used to unequivocally localize the stimulated synapse. This technique was developed to image, for the first time, the ultrastructure of the postsynaptic density in which long-term potentiation was selectively induced just seconds or minutes before, but it can be applied for the study of any biological process that requires the precise relocalization of micron-wide structures for their correlated imaging with 2PM and EM.
Collapse
Affiliation(s)
- Miquel Bosch
- RIKEN-MIT Neuroscience Research Center, Saitama, Japan.
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Universitat Internacional de Catalunya, Barcelona, Spain.
| | - Jorge Castro
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- MBF Bioscience, Williston, VT, USA
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yasunori Hayashi
- RIKEN-MIT Neuroscience Research Center, Saitama, Japan
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Brain Science Institute, RIKEN, Wako, Saitama, Japan
- Saitama University Brain Science Institute, Saitama University, Saitama, Japan
- School of Life Science, South China Normal University, Guangzhou, China
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
5
|
Jain A, Nakahata Y, Pancani T, Watabe T, Rusina P, South K, Adachi K, Yan L, Simorowski N, Furukawa H, Yasuda R. Dendritic, delayed, stochastic CaMKII activation in behavioural time scale plasticity. Nature 2024; 635:151-159. [PMID: 39385027 PMCID: PMC11540904 DOI: 10.1038/s41586-024-08021-8] [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: 08/08/2023] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Behavioural time scale plasticity (BTSP) is non-Hebbian plasticity induced by integrating presynaptic and postsynaptic components separated by a behaviourally relevant time scale (seconds)1. BTSP in hippocampal CA1 neurons underlies place cell formation. However, the molecular mechanisms that enable synapse-specific plasticity on a behavioural time scale are unknown. Here we show that BTSP can be induced in a single dendritic spine using two-photon glutamate uncaging paired with postsynaptic current injection temporally separated by a behavioural time scale. Using an improved Ca2+/calmodulin-dependent kinase II (CaMKII) sensor, we did not detect CaMKII activation during this BTSP induction. Instead, we observed dendritic, delayed and stochastic CaMKII activation (DDSC) associated with Ca2+ influx and plateau potentials 10-100 s after BTSP induction. DDSC required both presynaptic and postsynaptic activity, which suggests that CaMKII can integrate these two signals. Also, optogenetically blocking CaMKII 15-30 s after the BTSP protocol inhibited synaptic potentiation, which indicated that DDSC is an essential mechanism of BTSP. IP3-dependent intracellular Ca2+ release facilitated both DDSC and BTSP. Thus, our study suggests that non-synapse-specific CaMKII activation provides an instructive signal with an extensive time window over tens of seconds during BTSP.
Collapse
Affiliation(s)
- Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Centre for High Impact Neuroscience and Translational Applications (CHINTA), TCG CREST, Kolkata, India
| | - Yoshihisa Nakahata
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tristano Pancani
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tetsuya Watabe
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Polina Rusina
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kelly South
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kengo Adachi
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Long Yan
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
6
|
Huang S, Henderson TR, Dojo Soeandy C, Lezhanska A, Henderson JT. An efficient low cost means of biophysical gene transfection in primary cells. Sci Rep 2024; 14:13179. [PMID: 38849388 PMCID: PMC11161637 DOI: 10.1038/s41598-024-62996-y] [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: 10/30/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Efficient, facile gene modification of cells has become an indispensable part of modern molecular biology. For the majority of cell lines and several primary populations, such modifications can be readily performed through a variety of methods. However, many primary cell lines such as stem cells frequently suffer from poor transfection efficiency. Though several physical approaches have been introduced to circumvent these issues, they often require expensive/specialized equipment and/or consumables, utilize substantial cell numbers and often still suffer from poor efficiency. Viral methods are capable of transducing difficult cellular populations, however such methods can be time consuming for large arrays of gene targets, present biohazard concerns, and result in expression of viral proteins; issues of concern for certain experimental approaches. We report here a widely applicable, low-cost (< $100 CAD) method of electroporation, applicable to small (1-10 μl) cell volumes and composed of equipment readily available to the average investigator. Using this system we observe a sixfold increase in transfection efficiency in embryonic stem cell lines compared to commercial devices. Due to efficiency gains and reductions in volume and applied voltage, this process improves the survival of sensitive stem cell populations while reducing reagent requirements for protocols such as Cas9/gRNAs transfections.
Collapse
Affiliation(s)
- Shudi Huang
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada
| | - Tyler R Henderson
- Department of Medical Genetics, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Chesarahmia Dojo Soeandy
- Tumour Immunotherapy Program Cell Manufacturing Team, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Rm 8-207, Toronto, ON, M5G 2M9, Canada
| | - Anastasiya Lezhanska
- Department of Medicine, Queen's University, 94 Stuart Street, Kingston, ON, K7L 3N6, Canada
| | - Jeffrey T Henderson
- Department of Pharmaceutical Sciences, University of Toronto, 144 College St. Rm 962, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
7
|
Lu B, Lim JM, Yu B, Song S, Neeli P, Sobhani N, K P, Bonam SR, Kurapati R, Zheng J, Chai D. The next-generation DNA vaccine platforms and delivery systems: advances, challenges and prospects. Front Immunol 2024; 15:1332939. [PMID: 38361919 PMCID: PMC10867258 DOI: 10.3389/fimmu.2024.1332939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ming Lim
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Boyue Yu
- Department of Environmental Science, Policy, and Management, University of California at Berkeley, Berkeley, CA, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Pavithra K
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
8
|
Sembada AA, Lenggoro IW. Transport of Nanoparticles into Plants and Their Detection Methods. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:131. [PMID: 38251096 PMCID: PMC10819755 DOI: 10.3390/nano14020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Nanoparticle transport into plants is an evolving field of research with diverse applications in agriculture and biotechnology. This article provides an overview of the challenges and prospects associated with the transport of nanoparticles in plants, focusing on delivery methods and the detection of nanoparticles within plant tissues. Passive and assisted delivery methods, including the use of roots and leaves as introduction sites, are discussed, along with their respective advantages and limitations. The barriers encountered in nanoparticle delivery to plants are highlighted, emphasizing the need for innovative approaches (e.g., the stem as a new recognition site) to optimize transport efficiency. In recent years, research efforts have intensified, leading to an evendeeper understanding of the intricate mechanisms governing the interaction of nanomaterials with plant tissues and cells. Investigations into the uptake pathways and translocation mechanisms within plants have revealed nuanced responses to different types of nanoparticles. Additionally, this article delves into the importance of detection methods for studying nanoparticle localization and quantification within plant tissues. Various techniques are presented as valuable tools for comprehensively understanding nanoparticle-plant interactions. The reliance on multiple detection methods for data validation is emphasized to enhance the reliability of the research findings. The future outlooks of this field are explored, including the potential use of alternative introduction sites, such as stems, and the continued development of nanoparticle formulations that improve adhesion and penetration. By addressing these challenges and fostering multidisciplinary research, the field of nanoparticle transport in plants is poised to make significant contributions to sustainable agriculture and environmental management.
Collapse
Affiliation(s)
- Anca Awal Sembada
- Department of Applied Physics and Chemical Engineering, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
- School of Life Sciences and Technology, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - I. Wuled Lenggoro
- Department of Applied Physics and Chemical Engineering, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
| |
Collapse
|
9
|
Mavi AK, Kumar M, Singh A, Prajapati MK, Khabiya R, Maru S, Kumar D. Progress in Non‐Viral Delivery of Nucleic Acid. INTEGRATION OF BIOMATERIALS FOR GENE THERAPY 2023:281-322. [DOI: 10.1002/9781394175635.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Jain A, Nakahata Y, Watabe T, Rusina P, South K, Adachi K, Yan L, Simorowski N, Furukawa H, Yasuda R. Dendritic, delayed, and stochastic CaMKII activation underlies behavioral time scale plasticity in CA1 synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.549180. [PMID: 37577549 PMCID: PMC10418109 DOI: 10.1101/2023.08.01.549180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Behavioral time scale plasticity (BTSP), is a form of non-Hebbian plasticity induced by integrating pre- and postsynaptic components separated by behavioral time scale (seconds). BTSP in the hippocampal CA1 neurons underlies place cell formation. However, the molecular mechanisms underlying this behavioral time scale (eligibility trace) and synapse specificity are unknown. CaMKII can be activated in a synapse-specific manner and remain active for a few seconds, making it a compelling candidate for the eligibility trace during BTSP. Here, we show that BTSP can be induced in a single dendritic spine using 2-photon glutamate uncaging paired with postsynaptic current injection temporally separated by behavioral time scale. Using an improved CaMKII sensor, we saw no detectable CaMKII activation during this BTSP induction. Instead, we observed a dendritic, delayed, and stochastic CaMKII activation (DDSC) associated with Ca 2+ influx and plateau 20-40 s after BTSP induction. DDSC requires both pre-and postsynaptic activity, suggesting that CaMKII can integrate these two signals. Also, optogenetically blocking CaMKII 30 s after the BTSP protocol inhibited synaptic potentiation, indicating that DDSC is an essential mechanism of BTSP. IP3-dependent intracellular Ca 2+ release facilitates both DDSC and BTSP. Thus, our study suggests that the non-synapse specific CaMKII activation provides an instructive signal with an extensive time window over tens of seconds during BTSP.
Collapse
|
11
|
Colgan LA, Parra-Bueno P, Holman HL, Tu X, Jain A, Calubag MF, Misler JA, Gary C, Oz G, Suponitsky-Kroyter I, Okaz E, Yasuda R. Dual Regulation of Spine-Specific and Synapse-to-Nucleus Signaling by PKCδ during Plasticity. J Neurosci 2023; 43:5432-5447. [PMID: 37277178 PMCID: PMC10376934 DOI: 10.1523/jneurosci.0208-22.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023] Open
Abstract
The activity-dependent plasticity of synapses is believed to be the cellular basis of learning. These synaptic changes are mediated through the coordination of local biochemical reactions in synapses and changes in gene transcription in the nucleus to modulate neuronal circuits and behavior. The protein kinase C (PKC) family of isozymes has long been established as critical for synaptic plasticity. However, because of a lack of suitable isozyme-specific tools, the role of the novel subfamily of PKC isozymes is largely unknown. Here, through the development of fluorescence lifetime imaging-fluorescence resonance energy transfer activity sensors, we investigate novel PKC isozymes in synaptic plasticity in CA1 pyramidal neurons of mice of either sex. We find that PKCδ is activated downstream of TrkB and DAG production, and that the spatiotemporal nature of its activation depends on the plasticity stimulation. In response to single-spine plasticity, PKCδ is activated primarily in the stimulated spine and is required for local expression of plasticity. However, in response to multispine stimulation, a long-lasting and spreading activation of PKCδ scales with the number of spines stimulated and, by regulating cAMP response-element binding protein activity, couples spine plasticity to transcription in the nucleus. Thus, PKCδ plays a dual functional role in facilitating synaptic plasticity.SIGNIFICANCE STATEMENT Synaptic plasticity, or the ability to change the strength of the connections between neurons, underlies learning and memory and is critical for brain health. The protein kinase C (PKC) family is central to this process. However, understanding how these kinases work to mediate plasticity has been limited by a lack of tools to visualize and perturb their activity. Here, we introduce and use new tools to reveal a dual role for PKCδ in facilitating local synaptic plasticity and stabilizing this plasticity through spine-to-nucleus signaling to regulate transcription. This work provides new tools to overcome limitations in studying isozyme-specific PKC function and provides insight into molecular mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
- Lesley A Colgan
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Paula Parra-Bueno
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Heather L Holman
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Xun Tu
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Anant Jain
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Mariah F Calubag
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Jaime A Misler
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Chancellor Gary
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Goksu Oz
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Irena Suponitsky-Kroyter
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Elwy Okaz
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Ryohei Yasuda
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| |
Collapse
|
12
|
Kimura M, Endo A, Nagira Y, Yoshizumi T. Particle bombardment-assisted peptide-mediated gene transfer for highly efficient transient assay. BMC Res Notes 2023; 16:46. [PMID: 37024962 PMCID: PMC10080836 DOI: 10.1186/s13104-023-06320-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
OBJECTIVE A centrifugation-assisted peptide-mediated gene transfer (CAPT) method was recently developed as an efficient system for gene delivery into plant cells. However, the gene transfer efficiency of CAPT into plant cells was not entirely satisfactory for detecting transient expression of a transgene driven into mitochondria. Here, we report a new gene delivery system using a method called particle bombardment-assisted peptide-mediated gene transfer (PBPT). RESULTS We investigated various parameters of the PBPT method to increase transient gene expression efficiency in Brassica campestris. The optimal conditions for PBPT were a single bombardment with gold particles coated with a DNA‒peptide complex (6 µg of DNA and 2 µg of peptide) at an acceleration pressure of 5 kg/cm2 and a target distance of 12.5 cm. Moreover, bombardment under the optimal conditions successfully transferred the transgene into the cells of other plant species, namely B. juncea and tomato. Thus, we developed a PBPT method for highly efficient delivery of a DNA‒peptide complex into plant mitochondria.
Collapse
Affiliation(s)
- Mitsuhiro Kimura
- Faculty of Agriculture, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan.
- Faculty of Agriculture, Kyushu University, Fukuoka, Japan.
| | - Akira Endo
- Agri-Bio Research Center, Agri-Bio & Supplement Research Laboratories, Kaneka Corporation, Iwata, Shizuoka, Japan
| | - Yozo Nagira
- Agri-Bio Research Center, Agri-Bio & Supplement Research Laboratories, Kaneka Corporation, Iwata, Shizuoka, Japan
| | - Takeshi Yoshizumi
- Faculty of Agriculture, Takasaki University of Health and Welfare, Takasaki-shi, Gunma, Japan.
| |
Collapse
|
13
|
Wijesundara YH, Herbert FC, Trashi O, Trashi I, Brohlin OR, Kumari S, Howlett T, Benjamin CE, Shahrivarkevishahi A, Diwakara SD, Perera SD, Cornelius SA, Vizuet JP, Balkus KJ, Smaldone RA, De Nisco NJ, Gassensmith JJ. Carrier gas triggered controlled biolistic delivery of DNA and protein therapeutics from metal-organic frameworks. Chem Sci 2022; 13:13803-13814. [PMID: 36544734 PMCID: PMC9710232 DOI: 10.1039/d2sc04982a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/24/2022] [Indexed: 12/24/2022] Open
Abstract
The efficacy and specificity of protein, DNA, and RNA-based drugs make them popular in the clinic; however, these drugs are often delivered via injection, requiring skilled medical personnel, and producing biohazardous waste. Here, we report an approach that allows for their controlled delivery, affording either a burst or slow release without altering the formulation. We show that when encapsulated within zeolitic-imidazolate framework eight (ZIF-8), the biomolecules are stable in powder formulations and can be inoculated with a low-cost, gas-powered "MOF-Jet" into living animal and plant tissues. Additionally, their release profiles can be modulated through judicious selection of the carrier gas used in the MOF-Jet. Our in vitro and in vivo studies reveal that when CO2 is used, it creates a transient and weakly acidic local environment that causes a near-instantaneous release of the biomolecules through an immediate dissolution of ZIF-8. Conversely, when air is used, ZIF-8 biodegrades slowly, releasing the biomolecules over a week. This is the first example of controlled-biolistic delivery of biomolecules using ZIF-8, which provides a powerful tool for fundamental and applied science research.
Collapse
Affiliation(s)
- Yalini H. Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Fabian C. Herbert
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Orikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Ikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Olivia R. Brohlin
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Sneha Kumari
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Thomas Howlett
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Candace E. Benjamin
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Arezoo Shahrivarkevishahi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Shashini D. Diwakara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Sachini D. Perera
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Samuel A. Cornelius
- Department of Biological Sciences, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Juan P. Vizuet
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Kenneth J. Balkus
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Ronald A. Smaldone
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Nicole J. De Nisco
- Department of Biological Sciences, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA,Department of Biomedical Engineering, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| |
Collapse
|
14
|
Karpińska A, Zgorzelska A, Kwapiszewska K, Hołyst R. Entanglement of polymer chains in hypertonic medium enhances the delivery of DNA and other biomacromolecules into cells. J Colloid Interface Sci 2022; 627:270-282. [PMID: 35849860 DOI: 10.1016/j.jcis.2022.07.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022]
Abstract
HYPOTHESIS Most experimental procedures applied in modern biology involve cargo delivering into cells. One of the ways to cargo introduction is osmotic-mediated intracellular vesicle swelling. However, its widespread use was hindered due to cargo size (<10 nm) and cell-type-related restrictions. We addressed the issue of the composition of colloidal loading solution to enhance the efficiency of cellular delivery. EXPERIMENTS We examined the effectiveness of colloidal loading solutions of varied compositions, including various types and sizes of polymers building osmotic pressure. We used confocal imaging coupled with fluorescence correlation spectroscopy to evaluate the introduction of polymers, proteins, nanoparticles, and DNA plasmids (cargos of sizes 1-175 nm) to cells representing eight cell lines: cancer, normal, epithelial, and mesenchymal ones. FINDINGS We found that cellular delivery effectiveness strongly correlates with the size and concentration of osmotic pressure building polymers and not with the high value of the osmotic pressure itself. We show that polymer solutions at the entangled regime of concentrations enhance the delivery of large biomacromolecules even of size 200 nm (DNA plasmids) into cells, including MDA-MB-231 cells - so far resistant to the osmotic procedure. We show that the colloid loading medium based on entangled polymer chains is a versatile cargo delivery tool for molecular biology.
Collapse
Affiliation(s)
- Aneta Karpińska
- Department of Soft Condensed Matter, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| | - Alicja Zgorzelska
- Department of Soft Condensed Matter, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| | - Karina Kwapiszewska
- Department of Soft Condensed Matter, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| | - Robert Hołyst
- Department of Soft Condensed Matter, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland.
| |
Collapse
|
15
|
Topography-Mediated Enhancement of Nonviral Gene Delivery in Stem Cells. Pharmaceutics 2022; 14:pharmaceutics14051096. [PMID: 35631682 PMCID: PMC9142897 DOI: 10.3390/pharmaceutics14051096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/09/2022] [Accepted: 05/18/2022] [Indexed: 02/07/2023] Open
Abstract
Gene delivery holds great promise for bioengineering, biomedical applications, biosensors, diagnoses, and gene therapy. In particular, the influence of topography on gene delivery is considered to be an attractive approach due to low toxicity and localized delivery properties. Even though many gene vectors and transfection systems have been developed to enhance transfection potential and combining it with other forms of stimulations could even further enhance it. Topography is an interesting surface property that has been shown to stimulate differentiation, migration, cell morphology, and cell mechanics. Therefore, it is envisioned that topography might also be able to stimulate transfection. In this study, we tested the hypothesis “topography is able to regulate transfection efficiency”, for which we used nano- and microwave-like topographical substrates with wavelengths ranging from 500 nm to 25 µm and assessed the transfectability of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and myoblasts. For transfection, Lipofectamine 2000 and a gene encoding plasmid for red-fluorescent protein (m-Cherry) were used and topography-induced cell morphology and transfection efficiency was analyzed. As a result, topography directs cell spreading, elongation, and proliferation as well as the transfection efficiency, which were investigated but were found not to be correlated and dependent on the cell type. A 55% percent improvement of transfection efficiency was identified for hBM-MSCs grown on 2 µm wrinkles (24.3%) as compared to hBM-MSCs cultured on flat controls (15.7%). For myoblast cells, the highest gene-expression efficiency (46.1%) was observed on the 10 µm topography, which enhanced the transfection efficiency by 64% as compared to the flat control (28.1%). From a qualitative assessment, it was observed that the uptake capacity of cationic complexes of TAMRA-labeled oligodeoxynucleotides (ODNs) was not topography-dependent but that the intracellular release was faster, as indicated by the positively stained nuclei on 2 μm for hBM-MSCs and 10 μm for myoblasts. The presented results indicate that topography enhances the gene-delivery capacity and that the responses are dependent on cell type. This study demonstrates the important role of topography on cell stimulation for gene delivery as well as understanding the uptake capacity of lipoplexes and may be useful for developing advanced nonviral gene delivery strategies.
Collapse
|
16
|
Light triggered nanoscale biolistics for efficient intracellular delivery of functional macromolecules in mammalian cells. Nat Commun 2022; 13:1996. [PMID: 35422038 PMCID: PMC9010410 DOI: 10.1038/s41467-022-29713-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Biolistic intracellular delivery of functional macromolecules makes use of dense microparticles which are ballistically fired onto cells with a pressurized gun. While it has been used to transfect plant cells, its application to mammalian cells has met with limited success mainly due to high toxicity. Here we present a more refined nanotechnological approach to biolistic delivery with light-triggered self-assembled nanobombs (NBs) that consist of a photothermal core particle surrounded by smaller nanoprojectiles. Upon irradiation with pulsed laser light, fast heating of the core particle results in vapor bubble formation, which propels the nanoprojectiles through the cell membrane of nearby cells. We show successful transfection of both adherent and non-adherent cells with mRNA and pDNA, outperforming electroporation as the most used physical transfection technology by a factor of 5.5–7.6 in transfection yield. With a throughput of 104-105 cells per second, biolistic delivery with NBs offers scalable and highly efficient transfections of mammalian cells. Ballistic delivery with micro/nano-particles has been successfully used to transfect plant cells, however, has failed in mammalian cells due to toxic effects. Here, the authors report on a self-assembled nano-ballistic delivery system for the delivery of functional macromolecules and demonstrate efficient transfection of mammalian cells.
Collapse
|
17
|
Rajkumari N, Alex S, Soni KB, Anith KN, Viji MM, Kiran AG. Silver nanoparticles for biolistic transformation in Nicotiana tabacum L. 3 Biotech 2021; 11:497. [PMID: 34881160 DOI: 10.1007/s13205-021-03043-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022] Open
Abstract
The present study reports the use of silver nanoparticles as a gene carrier, substituting gold microcarrier for biolistic gene delivery in Nicotiana tabacum L. Efficiency of biolistic transformation using silver nanoparticles (100 nm) was compared with that of gold microcarriers (0.6 micron) under varying helium pressure (450 psi, 650 psi, 900 psi and 1100 psi) and target distance (6 cm and 9 cm). Among the different concentrations (0.01-100 mgL-1) of silver nanoparticles tried, 10 mgL-1 produced the highest number of transient GUS expression (30) with statistical significance. Helium pressure of 650 and target distance of 9 cm, and 900 psi pressure and 6 cm distance resulted in the highest GUS expression with gold microcarriers and silver nanoparticles, respectively. Transformation efficiency was significantly higher with silver nanoparticles than gold microparticles as carriers resulting in a reduction up to 37.5-fold on the cost of consumables. Regeneration efficiencies of tissues bombarded with gold microcarriers and silver nanoparticles were 62.5% and 70.83%, respectively.
Collapse
|
18
|
Using a Hand-Held Gene Gun for Genetic Transformation of Tetrahymena thermophila. Methods Mol Biol 2021. [PMID: 34542863 DOI: 10.1007/978-1-0716-1661-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Biolistic bombardment is widely used as a means of delivering vector-coated microparticles into microorganisms, cultured cells, and tissues. The first particle delivery system contained a helium propulsion unit (the gun) mounted in a vacuum-controlled chamber. In contrast, the hand-held gene gun does not operate within a chamber. It is completely hand-held, easy, and efficient to use, and it requires minimal space on the laboratory bench top. This chapter describes protocols for using a hand-held gene gun to deliver transformation vectors for overexpression of genes or gene replacement into the macronucleus of Tetrahymena thermophila. The protocols provide helpful information for preparing Tetrahymena for biolistic bombardment, preparation of vector-coated microcarriers, and basic gene gun operating procedures.
Collapse
|
19
|
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
|
20
|
Bodea SV, Westmeyer GG. Photoacoustic Neuroimaging - Perspectives on a Maturing Imaging Technique and its Applications in Neuroscience. Front Neurosci 2021; 15:655247. [PMID: 34220420 PMCID: PMC8253050 DOI: 10.3389/fnins.2021.655247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/08/2021] [Indexed: 11/13/2022] Open
Abstract
A prominent goal of neuroscience is to improve our understanding of how brain structure and activity interact to produce perception, emotion, behavior, and cognition. The brain's network activity is inherently organized in distinct spatiotemporal patterns that span scales from nanometer-sized synapses to meter-long nerve fibers and millisecond intervals between electrical signals to decades of memory storage. There is currently no single imaging method that alone can provide all the relevant information, but intelligent combinations of complementary techniques can be effective. Here, we thus present the latest advances in biomedical and biological engineering on photoacoustic neuroimaging in the context of complementary imaging techniques. A particular focus is placed on recent advances in whole-brain photoacoustic imaging in rodent models and its influential role in bridging the gap between fluorescence microscopy and more non-invasive techniques such as magnetic resonance imaging (MRI). We consider current strategies to address persistent challenges, particularly in developing molecular contrast agents, and conclude with an overview of potential future directions for photoacoustic neuroimaging to provide deeper insights into healthy and pathological brain processes.
Collapse
Affiliation(s)
- Silviu-Vasile Bodea
- Department of Chemistry and School of Medicine, Technical University of Munich (TUM), Munich, Germany
- Institute for Synthetic Biomedicine, Helmholtz Center Munich, Munich, Germany
| | - Gil Gregor Westmeyer
- Department of Chemistry and School of Medicine, Technical University of Munich (TUM), Munich, Germany
- Institute for Synthetic Biomedicine, Helmholtz Center Munich, Munich, Germany
| |
Collapse
|
21
|
Nanamiya R, Takei J, Asano T, Tanaka T, Sano M, Nakamura T, Yanaka M, Hosono H, Kaneko MK, Kato Y. Development of Anti-Human CC Chemokine Receptor 9 Monoclonal Antibodies for Flow Cytometry. Monoclon Antib Immunodiagn Immunother 2021; 40:101-106. [PMID: 34161159 DOI: 10.1089/mab.2021.0007] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
CC chemokine receptor 9 (CCR9) belongs to the beta chemokine receptor family and is mainly distributed on the surface of immature T lymphocytes and enterocytes. This receptor is highly expressed in rheumatoid arthritis, colitis, type 2 diabetes, and various tumors. Therefore, more sensitive monoclonal antibodies (mAbs) need to be developed to predict the prognosis of many high CCR9 expression diseases. Because CCR9 is a structurally unstable G protein-coupled receptor, it has been difficult to develop anti-CCR9 mAbs using the traditional method. This study developed anti-human CCR9 (hCCR9) mAbs for flow cytometry using a Cell-Based Immunization and Screening (CBIS) method. Two mice were immunized with hCCR9-overexpressed Chinese hamster ovary (CHO)-K1 cells (CHO/hCCR9), and hybridomas showing strong signals from CHO/hCCR9 and no signals from CHO-K1 cells were selected by flow cytometry. We established an anti-hCCR9 mAb, C9Mab-1 (IgG1, kappa), which detected hCCR9 in MOLT-4 leukemia T lymphoblast cells and CHO/hCCR9 cells by flow cytometry. Our study showed that an anti-hCCR9 mAb was developed more rapidly by the CBIS method than the previous method.
Collapse
Affiliation(s)
- Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Hosono
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| |
Collapse
|
22
|
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
|
23
|
Ai X, Wang S, Duan Y, Zhang Q, Chen M, Gao W, Zhang L. Emerging Approaches to Functionalizing Cell Membrane-Coated Nanoparticles. Biochemistry 2021; 60:941-955. [PMID: 32452667 PMCID: PMC8507422 DOI: 10.1021/acs.biochem.0c00343] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
There has been significant interest in developing cell membrane-coated nanoparticles due to their unique abilities of biomimicry and biointerfacing. As the technology progresses, it becomes clear that the application of these nanoparticles can be drastically broadened if additional functions beyond those derived from the natural cell membranes can be integrated. Herein, we summarize the most recent advances in the functionalization of cell membrane-coated nanoparticles. In particular, we focus on emerging methods, including (1) lipid insertion, (2) membrane hybridization, (3) metabolic engineering, and (4) genetic modification. These approaches contribute diverse functions in a nondisruptive fashion while preserving the natural function of the cell membranes. They also improve on the multifunctional and multitasking ability of cell membrane-coated nanoparticles, making them more adaptive to the complexity of biological systems. We hope that these approaches will serve as inspiration for more strategies and innovations to advance cell membrane coating technology.
Collapse
Affiliation(s)
- Xiangzhao Ai
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Shuyan Wang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Yaou Duan
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Qiangzhe Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Maggie Chen
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Weiwei Gao
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Liangfang Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
24
|
Grinman E, Nakahata Y, Avchalumov Y, Espadas I, Swarnkar S, Yasuda R, Puthanveettil SV. Activity-regulated synaptic targeting of lncRNA ADEPTR mediates structural plasticity by localizing Sptn1 and AnkB in dendrites. SCIENCE ADVANCES 2021; 7:7/16/eabf0605. [PMID: 33863727 PMCID: PMC8051873 DOI: 10.1126/sciadv.abf0605] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/26/2021] [Indexed: 05/26/2023]
Abstract
Activity-dependent structural plasticity at the synapse requires specific changes in the neuronal transcriptome. While much is known about the role of coding elements in this process, the role of the long noncoding transcriptome remains elusive. Here, we report the discovery of an intronic long noncoding RNA (lncRNA)-termed ADEPTR-that is up-regulated and synaptically transported in a cAMP/PKA-dependent manner in hippocampal neurons, independently of its protein-coding host gene. Loss of ADEPTR function suppresses activity-dependent changes in synaptic transmission and structural plasticity of dendritic spines. Mechanistically, dendritic localization of ADEPTR is mediated by molecular motor protein Kif2A. ADEPTR physically binds to actin-scaffolding regulators ankyrin (AnkB) and spectrin (Sptn1) via a conserved sequence and is required for their dendritic localization. Together, this study demonstrates how activity-dependent synaptic targeting of an lncRNA mediates structural plasticity at the synapse.
Collapse
Affiliation(s)
- Eddie Grinman
- Department of Neuroscience, Scripps Research, 130 Scripps Way, Jupiter, FL 33458, USA
| | | | - Yosef Avchalumov
- Department of Neuroscience, Scripps Research, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Isabel Espadas
- Department of Neuroscience, Scripps Research, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Supriya Swarnkar
- Department of Neuroscience, Scripps Research, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | | |
Collapse
|
25
|
Hasan MM, Ragnarsson L, Cardoso FC, Lewis RJ. Transfection methods for high-throughput cellular assays of voltage-gated calcium and sodium channels involved in pain. PLoS One 2021; 16:e0243645. [PMID: 33667217 PMCID: PMC7935312 DOI: 10.1371/journal.pone.0243645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Abstract
Chemical transfection is broadly used to transiently transfect mammalian cells, although often associated with cellular stress and membrane instability, which imposes challenges for most cellular assays, including high-throughput (HT) assays. In the current study, we compared the effectiveness of calcium phosphate, FuGENE and Lipofectamine 3000 to transiently express two key voltage-gated ion channels critical in pain pathways, CaV2.2 and NaV1.7. The expression and function of these channels were validated using two HT platforms, the Fluorescence Imaging Plate Reader FLIPRTetra and the automated patch clamp QPatch 16X. We found that all transfection methods tested demonstrated similar effectiveness when applied to FLIPRTetra assays. Lipofectamine 3000-mediated transfection produced the largest peak currents for automated patch clamp QPatch assays. However, the FuGENE-mediated transfection was the most effective for QPatch assays as indicated by the superior number of cells displaying GΩ seal formation in whole-cell patch clamp configuration, medium to large peak currents, and higher rates of accomplished assays for both CaV2.2 and NaV1.7 channels. Our findings can facilitate the development of HT automated patch clamp assays for the discovery and characterization of novel analgesics and modulators of pain pathways, as well as assisting studies examining the pharmacology of mutated channels.
Collapse
Affiliation(s)
- Md. Mahadhi Hasan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Fernanda C. Cardoso
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
- * E-mail: (FCC); (RJL)
| |
Collapse
|
26
|
Preparation of Rat Organotypic Hippocampal Slice Cultures Using the Membrane-Interface Method. Methods Mol Biol 2021; 2188:243-257. [PMID: 33119855 DOI: 10.1007/978-1-0716-0818-0_12] [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] [Indexed: 01/27/2023]
Abstract
Cultured hippocampal slices from rodents, in which the architecture and functional properties of the hippocampal network are largely preserved, have proved to be a powerful substrate for studying healthy and pathological neuronal mechanisms. Here, we delineate the membrane-interface method for maintaining organotypic slices in culture for several weeks. The protocol includes procedures for dissecting hippocampus from rat brain, and collecting slices using a vibratome. This method provides the experimenter with easy access to both the brain tissue and culture medium, which facilitates genetic and pharmacological manipulations and enables experiments that incorporate imaging and electrophysiology. The method is generally applicable to rats of different ages, and to different brain regions, and can be modified for culture of slices from other species including mice.
Collapse
|
27
|
Gantenbein B, Tang S, Guerrero J, Higuita-Castro N, Salazar-Puerta AI, Croft AS, Gazdhar A, Purmessur D. Non-viral Gene Delivery Methods for Bone and Joints. Front Bioeng Biotechnol 2020; 8:598466. [PMID: 33330428 PMCID: PMC7711090 DOI: 10.3389/fbioe.2020.598466] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Viral carrier transport efficiency of gene delivery is high, depending on the type of vector. However, viral delivery poses significant safety concerns such as inefficient/unpredictable reprogramming outcomes, genomic integration, as well as unwarranted immune responses and toxicity. Thus, non-viral gene delivery methods are more feasible for translation as these allow safer delivery of genes and can modulate gene expression transiently both in vivo, ex vivo, and in vitro. Based on current studies, the efficiency of these technologies appears to be more limited, but they are appealing for clinical translation. This review presents a summary of recent advancements in orthopedics, where primarily bone and joints from the musculoskeletal apparatus were targeted. In connective tissues, which are known to have a poor healing capacity, and have a relatively low cell-density, i.e., articular cartilage, bone, and the intervertebral disk (IVD) several approaches have recently been undertaken. We provide a brief overview of the existing technologies, using nano-spheres/engineered vesicles, lipofection, and in vivo electroporation. Here, delivery for microRNA (miRNA), and silencing RNA (siRNA) and DNA plasmids will be discussed. Recent studies will be summarized that aimed to improve regeneration of these tissues, involving the delivery of bone morphogenic proteins (BMPs), such as BMP2 for improvement of bone healing. For articular cartilage/osteochondral junction, non-viral methods concentrate on targeted delivery to chondrocytes or MSCs for tissue engineering-based approaches. For the IVD, growth factors such as GDF5 or GDF6 or developmental transcription factors such as Brachyury or FOXF1 seem to be of high clinical interest. However, the most efficient method of gene transfer is still elusive, as several preclinical studies have reported many different non-viral methods and clinical translation of these techniques still needs to be validated. Here we discuss the non-viral methods applied for bone and joint and propose methods that can be promising in clinical use.
Collapse
Affiliation(s)
- Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shirley Tang
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Julien Guerrero
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Andreas S Croft
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Devina Purmessur
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
28
|
Santra TS, Kar S, Chang HY, Tseng FG. Nano-localized single-cell nano-electroporation. LAB ON A CHIP 2020; 20:4194-4204. [PMID: 33047768 DOI: 10.1039/d0lc00712a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The ability to deliver foreign cargos into single living cells is of great interest in cell biology and therapeutic research. Here, we have reported a single or multiple position based nano-localized single-cell nano-electroporation platform. The device consists of an array of triangular shape ITO nano-electrodes with a 70 nm gap between two nano-electrodes, each having a 40 nm tip diameter. The voltage is applied between nano-electrodes to generate an intense electric field, which electroporates multiple nano-localized regions of the targeted single-cell membrane, and biomolecules are gently delivered into cells by pressurizing pump flow, without affecting cell viability. The platform successfully delivers dyes, QDs, and plasmids into different cell types with the variation of field strength, pulse duration, and the number of pulses. This new approach allows us to analyze delivery of different biomolecules into single living cells with high transfection efficiency (>96%, for CL1-0 cells) and high cell viability (∼98%), which are potentially beneficial for cellular therapy and diagnostic purposes.
Collapse
Affiliation(s)
- Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 60036, India.
| | - Srabani Kar
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 60036, India. and Electrical Engineering, University of Cambridge, CB3 0FA, UK
| | - Hwan-You Chang
- Department of Life Science, National Tsing Hua University, Hsinchu 30012, Taiwan and Department of Medical Science, National Tsing Hua University, Hsinchu 30012, Taiwan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30012, Taiwan and Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan and Frontier Research Center of Fundamental and Applied Sciences, National Tsing Hua University, Hsinchu 30012, Taiwan
| |
Collapse
|
29
|
Hamad MIK, Daoud S, Petrova P, Rabaya O, Jbara A, Melliti N, Stichmann S, Reiss G, Herz J, Förster E. Biolistic transfection and expression analysis of acute cortical slices. J Neurosci Methods 2020; 337:108666. [PMID: 32119875 DOI: 10.1016/j.jneumeth.2020.108666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Biolistic gene gun transfection has been used to transfect organotypic cultures (OTCs) or dissociated cultures in vitro. Here, we modified this technique to allow successful transfection of acute brain slices, followed by measurement of neuronal activity within a few hours. NEW METHOD We established biolistic transfection of murine acute cortical slices to measure calcium signals. Acute slices are mounted on plasma/thrombin coagulate and transfected with a calcium sensor. Imaging can be performed within 4 h post transfection without affecting cell viability. RESULTS Four hours after GCaMP6s transfection, acute slices display remarkable fluorescent protein expression level allowing to study spontaneous activity and receptor pharmacology. While optimal gas pressure (150 psi) and gold particle size used (1 μm) confirm previously published protocols, the amount of 5 μg DNA was found to be optimal for particle coating. COMPARISON WITH EXISTING METHODS The major advantage of this technique is the rapid disposition of acute slices for calcium imaging. No transgenic GECI expressing animals or OTC for long periods are required. In acute slices, network interaction and connectivity are preserved. The method allows to obtain physiological readouts within 4 h, before functional tissue modifications might come into effect. Limitations of this technique are random transfection, low expression efficiency when using specific promotors, and preclusion or genetic manipulations that require a prolonged time before physiological changes become measurable, such as expression of recombinant proteins that require transport to distant subcellular localizations. CONCLUSION The method is optimal for short-time investigation of calcium signals in acute slices.
Collapse
Affiliation(s)
- Mohammad I K Hamad
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany; Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany.
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Nesrine Melliti
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| | - Sarah Stichmann
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany
| |
Collapse
|
30
|
Non-Viral in Vitro Gene Delivery: It is Now Time to Set the Bar! Pharmaceutics 2020; 12:pharmaceutics12020183. [PMID: 32098191 PMCID: PMC7076396 DOI: 10.3390/pharmaceutics12020183] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/31/2023] Open
Abstract
Transfection by means of non-viral gene delivery vectors is the cornerstone of modern gene delivery. Despite the resources poured into the development of ever more effective transfectants, improvement is still slow and limited. Of note, the performance of any gene delivery vector in vitro is strictly dependent on several experimental conditions specific to each laboratory. The lack of standard tests has thus largely contributed to the flood of inconsistent data underpinning the reproducibility crisis. A way researchers seek to address this issue is by gauging the effectiveness of newly synthesized gene delivery vectors with respect to benchmarks of seemingly well-known behavior. However, the performance of such reference molecules is also affected by the testing conditions. This survey points to non-standardized transfection settings and limited information on variables deemed relevant in this context as the major cause of such misalignments. This review provides a catalog of conditions optimized for the gold standard and internal reference, 25 kDa polyethyleneimine, that can be profitably replicated across studies for the sake of comparison. Overall, we wish to pave the way for the implementation of standardized protocols in order to make the evaluation of the effectiveness of transfectants as unbiased as possible.
Collapse
|
31
|
Tu X, Yasuda R, Colgan LA. Rac1 is a downstream effector of PKCα in structural synaptic plasticity. Sci Rep 2020; 10:1777. [PMID: 32019972 PMCID: PMC7000694 DOI: 10.1038/s41598-020-58610-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/17/2020] [Indexed: 11/21/2022] Open
Abstract
Structural and functional plasticity of dendritic spines is the basis of animal learning. The rapid remodeling of actin cytoskeleton is associated with spine enlargement and shrinkage, which are essential for structural plasticity. The calcium-dependent protein kinase C isoform, PKCα, has been suggested to be critical for this actin-dependent plasticity. However, mechanisms linking PKCα and structural plasticity of spines are unknown. Here, we examine the spatiotemporal activation of actin regulators, including small GTPases Rac1, Cdc42 and Ras, in the presence or absence of PKCα during single-spine structural plasticity. Removal of PKCα expression in the postsynapse attenuated Rac1 activation during structural plasticity without affecting Ras or Cdc42 activity. Moreover, disruption of a PDZ binding domain within PKCα led to impaired Rac1 activation and deficits in structural spine remodeling. These results demonstrate that PKCα positively regulates the activation of Rac1 during structural plasticity.
Collapse
Affiliation(s)
- Xun Tu
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
- International Max Planck Research School for Brain and Behavior, Jupiter, FL, USA.
- FAU/Max Planck Florida Institute Joint Graduate Program in Integrative Biology and Neuroscience, Florida Atlantic University, Boca Raton, FL, USA.
| | - Lesley A Colgan
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
32
|
Chen Z, Li H, Zhang L, Lee CK, Ho LWC, Chan CKW, Yang H, Choi CHJ. Specific Delivery of Oligonucleotides to the Cell Nucleus via Gentle Compression and Attachment of Polythymidine. ACS APPLIED MATERIALS & INTERFACES 2019; 11:27624-27640. [PMID: 31303000 DOI: 10.1021/acsami.9b11391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nonviral delivery of nucleic acids to the cell nucleus typically requires chemical methods that do not guarantee specific delivery (e.g., transfection agent) or physical methods that may require extensive fabrication (e.g., microfluidics) or an elevated pressure (e.g., 105 Pa for microneedles). We report a method of delivering oligonucleotides to the nucleus with high specificity (relative to the cytosol) by synergistically combining chemical and physical approaches. Particularly, we demonstrate that DNA oligonucleotides appended with a polythymidine [poly(T)] segment (chemical) profusely accumulate inside the nucleus when the cells are under gentle compression imposed by the weight of a single glass coverslip (physical; ∼2.2 Pa). Our "compression-cum-poly(T)" delivery method is simple, can be generalizable to three "hard-to-transfect" cell types, and does not induce significant levels of cytotoxicity or long-term oxidative stress to the treated cells when provided the use of suitable compression times and oligonucleotide concentrations. In bEnd.3 endothelial cells, compression-aided intranuclear delivery of poly(T) is primarily mediated by importin β and nucleoporin 62. Our method significantly enhances the intranuclear delivery of antisense oligonucleotides to bEnd.3 endothelioma cells and the inhibition of two target genes, including a reporter gene encoding the enhanced green fluorescent protein and an intranuclear lncRNA oncogene (metastasis-associated lung adenocarcinoma transcript 1), when compared with delivery without gentle compression or poly(T) attachment. Our data underscore the critical roles of pressure and nucleotide sequence on the intranuclear delivery of nucleic acids.
Collapse
|
33
|
Linsley JW, Tripathi A, Epstein I, Schmunk G, Mount E, Campioni M, Oza V, Barch M, Javaherian A, Nowakowski TJ, Samsi S, Finkbeiner S. Automated four-dimensional long term imaging enables single cell tracking within organotypic brain slices to study neurodevelopment and degeneration. Commun Biol 2019; 2:155. [PMID: 31069265 PMCID: PMC6494885 DOI: 10.1038/s42003-019-0411-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/18/2019] [Indexed: 02/08/2023] Open
Abstract
Current approaches for dynamic profiling of single cells rely on dissociated cultures, which lack important biological features existing in tissues. Organotypic slice cultures preserve aspects of structural and synaptic organisation within the brain and are amenable to microscopy, but established techniques are not well adapted for high throughput or longitudinal single cell analysis. Here we developed a custom-built, automated confocal imaging platform, with improved organotypic slice culture and maintenance. The approach enables fully automated image acquisition and four-dimensional tracking of morphological changes within individual cells in organotypic cultures from rodent and human primary tissues for at least 3 weeks. To validate this system, we analysed neurons expressing a disease-associated version of huntingtin (HTT586Q138-EGFP), and observed that they displayed hallmarks of Huntington's disease and died sooner than controls. By facilitating longitudinal single-cell analyses of neuronal physiology, our system bridges scales necessary to attain statistical power to detect developmental and disease phenotypes.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Atmiyata Tripathi
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Irina Epstein
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Galina Schmunk
- 2Department of Anatomy, University of California, San Francisco, CA 94158 USA
| | - Elliot Mount
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Matthew Campioni
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Viral Oza
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Mariya Barch
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Ashkan Javaherian
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
| | - Tomasz J Nowakowski
- 2Department of Anatomy, University of California, San Francisco, CA 94158 USA
| | - Siddharth Samsi
- 3Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, L-4367 Luxembourg
- 9Present Address: MIT Lincoln Laboratory, Lexington, MA 02421 USA
| | - Steven Finkbeiner
- Gladstone Center for Systems and Therapeutics, San Francisco, CA 94158 USA
- 4Neuroscience Graduate Program, University of California, San Francisco, CA 94158 USA
- 5Biomedical Sciences and Neuroscience Graduate Program, University of California, San Francisco, CA 94143 USA
- 6Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA 94158 USA
- 7Department of Neurology, University of California, San Francisco, CA 94158 USA
- 8Department of Physiology, University of California, San Francisco, CA 94158 USA
| |
Collapse
|
34
|
Acanda Y, Wang C, Levy A. Gene Expression in Citrus Plant Cells Using Helios ® Gene Gun System for Particle Bombardment. Methods Mol Biol 2019; 2015:219-228. [PMID: 31222707 DOI: 10.1007/978-1-4939-9558-5_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To understand how Citrus tristeza virus (CTV) replicates and moves inside the plant, it is critical to study the cellular interactions and localization of its encoded proteins. However, due to technical limitations, so far these studies have been limited to the nonnatural host Nicotiana benthamiana.Particle bombardment is a physical method to deliver nucleic acid and other biomolecules into the cells directly. The Helios® gene gun (Bio-Rad, Hercules, CA) is a handheld device that uses a low-pressure helium pulse to accelerate high-density, subcellular-sized particles into a wide variety of targets for in vivo and in vitro applications. Here, we describe a detail protocol for either transient or stable gene expression in citrus leaf cells using this gene gun. This protocol can be used to study protein-protein interactions and subcellular localization in different kinds of plant cells.
Collapse
Affiliation(s)
- Yosvanis Acanda
- Department of Plant Pathology, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Chunxia Wang
- Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA
| | - Amit Levy
- Department of Plant Pathology, Citrus Research and Education Center, University of Florida, Lake Alfred, FL, USA.
| |
Collapse
|
35
|
Hallin EI, Eriksen MS, Baryshnikov S, Nikolaienko O, Grødem S, Hosokawa T, Hayashi Y, Bramham CR, Kursula P. Structure of monomeric full-length ARC sheds light on molecular flexibility, protein interactions, and functional modalities. J Neurochem 2018; 147:323-343. [DOI: 10.1111/jnc.14556] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/29/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Erik I. Hallin
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - Maria S. Eriksen
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Sergei Baryshnikov
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Oleksii Nikolaienko
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Sverre Grødem
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Tomohisa Hosokawa
- Department of Pharmacology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Yasunori Hayashi
- Department of Pharmacology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Clive R. Bramham
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Petri Kursula
- Department of Biomedicine; University of Bergen; Bergen Norway
- Faculty of Biochemistry and Molecular Medicine; University of Oulu; Oulu Finland
| |
Collapse
|
36
|
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
|
37
|
Stewart MP, Langer R, Jensen KF. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem Rev 2018; 118:7409-7531. [PMID: 30052023 PMCID: PMC6763210 DOI: 10.1021/acs.chemrev.7b00678] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular delivery is a key step in biological research and has enabled decades of biomedical discoveries. It is also becoming increasingly important in industrial and medical applications ranging from biomanufacture to cell-based therapies. Here, we review techniques for membrane disruption-based intracellular delivery from 1911 until the present. These methods achieve rapid, direct, and universal delivery of almost any cargo molecule or material that can be dispersed in solution. We start by covering the motivations for intracellular delivery and the challenges associated with the different cargo types-small molecules, proteins/peptides, nucleic acids, synthetic nanomaterials, and large cargo. The review then presents a broad comparison of delivery strategies followed by an analysis of membrane disruption mechanisms and the biology of the cell response. We cover mechanical, electrical, thermal, optical, and chemical strategies of membrane disruption with a particular emphasis on their applications and challenges to implementation. Throughout, we highlight specific mechanisms of membrane disruption and suggest areas in need of further experimentation. We hope the concepts discussed in our review inspire scientists and engineers with further ideas to improve intracellular delivery.
Collapse
Affiliation(s)
- Martin P. Stewart
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
- The Koch Institute for Integrative Cancer Research,
Massachusetts Institute of Technology, Cambridge, USA
| | - Klavs F. Jensen
- Department of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, USA
| |
Collapse
|
38
|
PKCα integrates spatiotemporally distinct Ca 2+ and autocrine BDNF signaling to facilitate synaptic plasticity. Nat Neurosci 2018; 21:1027-1037. [PMID: 30013171 PMCID: PMC6100743 DOI: 10.1038/s41593-018-0184-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 05/29/2018] [Indexed: 11/24/2022]
Abstract
The Protein Kinase C (PKC) enzymes have long been established as critical for synaptic plasticity. However, it is unknown whether Ca2+-dependent PKC isozymes are activated in dendritic spines during plasticity, and if so, how this synaptic activity is encoded by PKC. Here, using newly-developed, isozyme-specific sensors, we demonstrate that classic isozymes are activated to varying degrees and with unique kinetics. PKCα is activated robustly and rapidly in stimulated spines and is the only isozyme required for structural plasticity. This specificity, depends on a PDZ-binding domain present only in PKCα. The activation of PKCα during plasticity requires both NMDAR Ca2+-flux and autocrine BDNF-TrkB signaling, two pathways that differ vastly in their spatiotemporal scales of signaling. Our results suggest that by integrating these signals, PKCα combines a measure of recent, nearby synaptic plasticity with local synaptic input, enabling complex cellular computations such as heterosynaptic facilitation of plasticity necessary for efficient hippocampal-dependent learning.
Collapse
|
39
|
Rajgor D, Sanderson TM, Amici M, Collingridge GL, Hanley JG. NMDAR-dependent Argonaute 2 phosphorylation regulates miRNA activity and dendritic spine plasticity. EMBO J 2018; 37:e97943. [PMID: 29712715 PMCID: PMC5983126 DOI: 10.15252/embj.201797943] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) repress translation of target mRNAs by associating with Argonaute (Ago) proteins to form the RNA-induced silencing complex (RISC), underpinning a powerful mechanism for fine-tuning protein expression. Specific miRNAs are required for NMDA receptor (NMDAR)-dependent synaptic plasticity by modulating the translation of proteins involved in dendritic spine morphogenesis or synaptic transmission. However, it is unknown how NMDAR stimulation stimulates RISC activity to rapidly repress translation of synaptic proteins. We show that NMDAR stimulation transiently increases Akt-dependent phosphorylation of Ago2 at S387, which causes an increase in binding to GW182 and a rapid increase in translational repression of LIMK1 via miR-134. Furthermore, NMDAR-dependent down-regulation of endogenous LIMK1 translation in dendrites and dendritic spine shrinkage requires phospho-regulation of Ago2 at S387. AMPAR trafficking and hippocampal LTD do not involve S387 phosphorylation, defining this mechanism as a specific pathway for structural plasticity. This work defines a novel mechanism for the rapid transduction of NMDAR stimulation into miRNA-mediated translational repression to control dendritic spine morphology.
Collapse
Affiliation(s)
- Dipen Rajgor
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, UK
| | - Thomas M Sanderson
- Centre for Synaptic Plasticity and School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Mascia Amici
- Centre for Synaptic Plasticity and School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Graham L Collingridge
- Centre for Synaptic Plasticity and School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, University of Bristol, Bristol, UK
| |
Collapse
|
40
|
López-Marín LM, Rivera AL, Fernández F, Loske AM. Shock wave-induced permeabilization of mammalian cells. Phys Life Rev 2018; 26-27:1-38. [PMID: 29685859 DOI: 10.1016/j.plrev.2018.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/26/2018] [Indexed: 12/18/2022]
Abstract
Controlled permeabilization of mammalian cell membranes is fundamental to develop gene and cell therapies based on macromolecular cargo delivery, a process that emerged against an increasing number of health afflictions, including genetic disorders, cancer and infections. Viral vectors have been successfully used for macromolecular delivery; however, they may have unpredictable side effects and have been limited to life-threatening cases. Thus, several chemical and physical methods have been explored to introduce drugs, vaccines, and nucleic acids into cells. One of the most appealing physical methods to deliver genes into cells is shock wave-induced poration. High-speed microjets of fluid, emitted due to the collapse of microbubbles after shock wave passage, represent the most significant mechanism that contributes to cell membrane poration by this technique. Herein, progress in shock wave-induced permeabilization of mammalian cells is presented. After covering the main concepts related to molecular strategies whose applications depend on safer drug delivery methods, the physics behind shock wave phenomena is described. Insights into the use of shock waves for cell membrane permeation are discussed, along with an overview of the two major biomedical applications thereof-i.e., genetic modification and anti-cancer shock wave-assisted chemotherapy. The aim of this review is to summarize 30 years of data showing underwater shock waves as a safe, noninvasive method for macromolecular delivery into mammalian cells, encouraging the development of further research, which is still required before the introduction of this promising tool into clinical practice.
Collapse
Affiliation(s)
- Luz M López-Marín
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| | - Ana Leonor Rivera
- Instituto de Ciencias Nucleares & Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Ciudad de México, Mexico.
| | - Francisco Fernández
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| | - Achim M Loske
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230 Querétaro, Qro., Mexico.
| |
Collapse
|
41
|
Cheng MA, Farmer E, Huang C, Lin J, Hung CF, Wu TC. Therapeutic DNA Vaccines for Human Papillomavirus and Associated Diseases. Hum Gene Ther 2018; 29:971-996. [PMID: 29316817 DOI: 10.1089/hum.2017.197] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human papillomavirus (HPV) has long been recognized as the causative agent of cervical cancer. High-risk HPV types 16 and 18 alone are responsible for over 70% of all cases of cervical cancers. More recently, HPV has been identified as an etiological factor for several other forms of cancers, including oropharyngeal, anogenital, and skin. Thus, the association of HPV with these malignancies creates an opportunity to control these HPV lesions and HPV-associated malignancies through immunization. Strategies to prevent or to therapeutically treat HPV infections have been developed and are still pushing innovative boundaries. Currently, commercial prophylactic HPV vaccines are widely available, but they are not able to control established infections or lesions. As a result, there is an urgent need for the development of therapeutic HPV vaccines, to treat existing infections, and to prevent the development of HPV-associated cancers. In particular, DNA vaccination has emerged as a promising form of therapeutic HPV vaccine. DNA vaccines have great potential for the treatment of HPV infections and HPV-associated cancers due to their safety, stability, simplicity of manufacturability, and ability to induce antigen-specific immunity. This review focuses on the current state of therapeutic HPV DNA vaccines, including results from recent and ongoing clinical trials, and outlines different strategies that have been employed to improve their potencies. The continued progress and improvements made in therapeutic HPV DNA vaccine development holds great potential for innovative ways to effectively treat HPV infections and HPV-associated diseases.
Collapse
Affiliation(s)
- Max A Cheng
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Emily Farmer
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Claire Huang
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - John Lin
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Chien-Fu Hung
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - T-C Wu
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,3 Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,4 Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| |
Collapse
|
42
|
Cornelia Koeberle S, Tanaka S, Kuriu T, Iwasaki H, Koeberle A, Schulz A, Helbing DL, Yamagata Y, Morrison H, Okabe S. Developmental stage-dependent regulation of spine formation by calcium-calmodulin-dependent protein kinase IIα and Rap1. Sci Rep 2017; 7:13409. [PMID: 29042611 PMCID: PMC5645322 DOI: 10.1038/s41598-017-13728-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/27/2017] [Indexed: 12/18/2022] Open
Abstract
The roles of calcium-calmodulin-dependent protein kinase II-alpha (CaMKIIα) in the expression of long-term synaptic plasticity in the adult brain have been extensively studied. However, how increased CaMKIIα activity controls the maturation of neuronal circuits remains incompletely understood. Herein, we show that pyramidal neurons without CaMKIIα activity upregulate the rate of spine addition, resulting in elevated spine density. Genetic elimination of CaMKIIα activity specifically eliminated the observed maturation-dependent suppression of spine formation. Enhanced spine formation was associated with the stabilization of actin in the spine and could be reversed by increasing the activity of the small GTPase Rap1. CaMKIIα activity was critical in the phosphorylation of synaptic Ras GTPase-activating protein (synGAP), the dispersion of synGAP from postsynaptic sites, and the activation of postsynaptic Rap1. CaMKIIα is already known to be essential in learning and memory, but our findings suggest that CaMKIIα plays an important activity-dependent role in restricting spine density during postnatal development.
Collapse
Affiliation(s)
- Solveigh Cornelia Koeberle
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Institute of Age Research, Fritz Lipmann Institute, Jena, Germany
| | - Shinji Tanaka
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,CREST, JST, Japan
| | - Toshihiko Kuriu
- Department of Neurophysiology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa, 769-2193, Japan
| | - Hirohide Iwasaki
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,CREST, JST, Japan
| | - Andreas Koeberle
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University, Jena, Germany
| | - Alexander Schulz
- Institute of Age Research, Fritz Lipmann Institute, Jena, Germany
| | | | - Yoko Yamagata
- Division of Neural Signaling, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan.,SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Helen Morrison
- Institute of Age Research, Fritz Lipmann Institute, Jena, Germany
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. .,CREST, JST, Japan.
| |
Collapse
|
43
|
Ding X, Stewart M, Sharei A, Weaver JC, Langer RS, Jensen KF. High-throughput Nuclear Delivery and Rapid Expression of DNA via Mechanical and Electrical Cell-Membrane Disruption. Nat Biomed Eng 2017; 1. [PMID: 28932622 PMCID: PMC5602535 DOI: 10.1038/s41551-017-0039] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Nuclear transfection of DNA into mammalian cells is challenging yet critical for many biological and medical studies. Here, by combining cell squeezing and electric-field-driven transport in a device that integrates microfluidic channels with constrictions and microelectrodes, we demonstrate nuclear delivery of plasmid DNA within 1 hour after treatment, the most rapid DNA expression in a high-throughput setting (up to millions of cells per minute per device). Passing cells at high speed through microfluidic constrictions smaller than the cell diameter mechanically disrupts the cell membrane, allowing a subsequent electric field to further disrupt the nuclear envelope and drive DNA molecules into the cytoplasm and nucleus. By tracking the localization of the ESCRT-III (endosomal sorting complexes required for transport) protein CHMP4B, we show that the integrity of the nuclear envelope is recovered within 15 minutes of treatment. We also provide insight into subcellular delivery by comparing the performance of the disruption-and-field-enhanced method with those of conventional chemical, electroporation, and manual-injection systems.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,The David Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Martin Stewart
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,The David Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Armon Sharei
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,The David Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James C Weaver
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert S Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,The David Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Klavs F Jensen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
44
|
Bosch M, Castro J, Sur M, Hayashi Y. Photomarking Relocalization Technique for Correlated Two-Photon and Electron Microcopy Imaging of Single Stimulated Synapses. Methods Mol Biol 2017; 1538:185-214. [PMID: 27943192 DOI: 10.1007/978-1-4939-6688-2_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Synapses learn and remember by persistent modifications of their internal structures and composition but, due to their small size, it is difficult to observe these changes at the ultrastructural level in real time. Two-photon fluorescence microscopy (2PM) allows time-course live imaging of individual synapses but lacks ultrastructural resolution. Electron microscopy (EM) allows the ultrastructural imaging of subcellular components but cannot detect fluorescence and lacks temporal resolution. Here, we describe a combination of procedures designed to achieve the correlated imaging of the same individual synapse under both 2PM and EM. This technique permits the selective stimulation and live imaging of a single dendritic spine and the subsequent localization of the same spine in EM ultrathin serial sections. Landmarks created through a photomarking method based on the 2-photon-induced precipitation of an electrodense compound are used to unequivocally localize the stimulated synapse. This technique was developed to image, for the first time, the ultrastructure of the postsynaptic density in which long-term potentiation was selectively induced just seconds or minutes before, but it can be applied for the study of any biological process that requires the precise relocalization of micron-wide structures for their correlated imaging with 2PM and EM.
Collapse
Affiliation(s)
- Miquel Bosch
- RIKEN-MIT Neuroscience Research Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Bioengineering of Catalonia, Barcelona, Spain.
| | - Jorge Castro
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yasunori Hayashi
- RIKEN-MIT Neuroscience Research Center, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Brain Science Institute, RIKEN, Wako, Saitama, Japan
- Saitama University Brain Science Institute, Saitama University, Saitama, Japan
- School of Life Science, South China Normal University, Guangzhou, China
- Department of Pharmacology, Faculty of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
45
|
Laviv T, Kim BB, Chu J, Lam AJ, Lin MZ, Yasuda R. Simultaneous dual-color fluorescence lifetime imaging with novel red-shifted fluorescent proteins. Nat Methods 2016; 13:989-992. [PMID: 27798609 PMCID: PMC5322478 DOI: 10.1038/nmeth.4046] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/30/2016] [Indexed: 12/18/2022]
Abstract
We describe a red-shifted fluorescence resonance energy transfer (FRET) pair optimized for dual-color fluorescence lifetime imaging (FLIM). This pair utilizes a newly developed FRET donor, monomeric cyan-excitable red fluorescent protein (mCyRFP1), which has a large Stokes shift and a monoexponential fluorescence lifetime decay. When used together with EGFP-based biosensors, the new pair enables simultaneous imaging of the activities of two signaling molecules in single dendritic spines undergoing structural plasticity.
Collapse
Affiliation(s)
- Tal Laviv
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| | - Benjamin B Kim
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Jun Chu
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Amy J Lam
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Michael Z Lin
- Department of Bioengineering, Stanford University, Stanford, California, USA
- Department of Pediatrics, Stanford University, Stanford, California, USA
- Department of Neurobiology, Stanford University, Stanford, California, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| |
Collapse
|
46
|
Golshadi M, Wright LK, Dickerson IM, Schrlau MG. High-Efficiency Gene Transfection of Cells through Carbon Nanotube Arrays. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3014-3020. [PMID: 27059518 DOI: 10.1002/smll.201503878] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/04/2016] [Indexed: 06/05/2023]
Abstract
Introducing nucleic acids into mammalian cells is a crucial step to elucidate biochemical pathways, and to modify gene expression and cellular development in immortalized cells, primary cells, and stem cells. Current transfection technologies are time consuming and limited by the size of genetic cargo, the inefficient introduction of test molecules into large populations of target cells, and the cytotoxicity of the techniques. A novel method of introducing genes and biomolecules into tens of thousands of mammalian cells has been developed through an array of aligned hollow carbon nanotubes, manufactured by template-based nanofabrication processes, to achieve rapid high-efficiency transfer with low cytotoxicity. The utilization of carbon nanotube arrays for gene transfection overcomes molecular weight limits of current technologies and can be adapted to deliver drugs or proteins in addition to nucleic acids.
Collapse
Affiliation(s)
- Masoud Golshadi
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Leslie K Wright
- School of Life Sciences, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Ian M Dickerson
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Michael G Schrlau
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| |
Collapse
|
47
|
Aps LRMM, Tavares MB, Rozenfeld JHK, Lamy MT, Ferreira LCS, Diniz MO. Bacterial spores as particulate carriers for gene gun delivery of plasmid DNA. J Biotechnol 2016; 228:58-66. [PMID: 27130499 DOI: 10.1016/j.jbiotec.2016.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022]
Abstract
Bacillus subtilis spores represent a suitable platform for the adsorption of proteins, enzymes and viral particles at physiological conditions. In the present work, we demonstrate that purified spores can also adsorb DNA on their surface after treatment with cationic molecules. In addition, we demonstrate that DNA-coated B. subtilis spores can be used as particulate carriers and act as an alternative to gold microparticles for the biolistic (gene gun) administration of plasmid DNA in mice. Gene gun delivery of spores pre-treated with DODAB (dioctadecyldimethylammonium bromide) allowed efficient plasmid DNA absorption and induced protein expression levels similar to those obtained with gold microparticles. More importantly, we demonstrated that a DNA vaccine adsorbed on spores can be loaded into biolistic cartridges and efficiently delivered into mice, which induced specific cellular and antibody responses. Altogether, these data indicate that B. subtilis spores represent a simple and low cost alternative for the in vivo delivery of DNA vaccines by the gene gun technology.
Collapse
Affiliation(s)
- Luana R M M Aps
- Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Milene B Tavares
- Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - M Teresa Lamy
- Institute of Physics, University of São Paulo, Brazil
| | - Luís C S Ferreira
- Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana O Diniz
- Vaccine Development Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
48
|
Berlin S, Szobota S, Reiner A, Carroll EC, Kienzler MA, Guyon A, Xiao T, Trauner D, Isacoff EY. A family of photoswitchable NMDA receptors. eLife 2016; 5. [PMID: 26929991 PMCID: PMC4786437 DOI: 10.7554/elife.12040] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/31/2016] [Indexed: 02/07/2023] Open
Abstract
NMDA receptors, which regulate synaptic strength and are implicated in learning and memory, consist of several subtypes with distinct subunit compositions and functional properties. To enable spatiotemporally defined, rapid and reproducible manipulation of function of specific subtypes, we engineered a set of photoswitchable GluN subunits ('LiGluNs'). Photo-agonism of GluN2A or GluN2B elicits an excitatory drive to hippocampal neurons that can be shaped in time to mimic synaptic activation. Photo-agonism of GluN2A at single dendritic spines evokes spine-specific calcium elevation and expansion, the morphological correlate of LTP. Photo-antagonism of GluN2A alone, or in combination with photo-antagonism of GluN1a, reversibly blocks excitatory synaptic currents, prevents the induction of long-term potentiation and prevents spine expansion. In addition, photo-antagonism in vivo disrupts synaptic pruning of developing retino-tectal projections in larval zebrafish. By providing precise and rapidly reversible optical control of NMDA receptor subtypes, LiGluNs should help unravel the contribution of specific NMDA receptors to synaptic transmission, integration and plasticity.
Collapse
Affiliation(s)
- Shai Berlin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Stephanie Szobota
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Andreas Reiner
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Elizabeth C Carroll
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Michael A Kienzler
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Alice Guyon
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia Antipolis, Nice, France
| | - Tong Xiao
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Dirk Trauner
- Department of Chemistry, Center of Integrated Protein Science, University of Munich, Munich, Germany
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, United States.,Physical Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| |
Collapse
|
49
|
Santra TS, Chen CW, Chang HY, Tseng FG. Dielectric passivation layer as a substratum on localized single-cell electroporation. RSC Adv 2016. [DOI: 10.1039/c5ra18258a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Without dielectric passivation layer provide bulk electroporation, whereas with passivation layer generate an intense electric field to deliver molecules precisely into single cell, as name as localized single cell electroporation (LSCEP).
Collapse
Affiliation(s)
- Tuhin Subhra Santra
- Institute of Nano Engineering and Microsystems
- National Tsing Hua University
- Taiwan
- Department of Engineering and System Science
- National Tsing Hua University
| | - Chih-Wei Chen
- Institute of Molecular Medicine
- National Tsing Hua University
- Taiwan
| | - Hwan-You Chang
- Institute of Molecular Medicine
- National Tsing Hua University
- Taiwan
| | - Fan-Gang Tseng
- Department of Engineering and System Science
- National Tsing Hua University
- Taiwan
- California Nano System Institute
- University of California at Los Angeles
| |
Collapse
|
50
|
Helios(®) Gene Gun-Mediated Transfection of the Inner Ear Sensory Epithelium: Recent Updates. Methods Mol Biol 2016; 1427:3-26. [PMID: 27259918 DOI: 10.1007/978-1-4939-3615-1_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The transfection of vertebrate inner ear hair cells has proven to be challenging. Therefore, many laboratories attempt to use and improve different transfection methods. Each method has its own advantages and disadvantages. A particular researcher's skills in addition to available equipment and the type of experiment (in vivo or in vitro) likely determine the transfection method of choice. Biolistic delivery of exogenous DNA, mRNA, or siRNA, also known as Helios(®) Gene Gun-mediated transfection, uses the mechanical energy of compressed helium gas to bombard tissue with micron- or submicron-sized DNA or RNA-coated gold particles, which can penetrate and transfect cells in vitro or in vivo. Helios(®) Gene Gun-mediated transfection has several advantages: (1) it is simple enough to learn in a short time; (2) it is designed to overcome cell barriers even as tough as plant cell membrane or stratum corneum in the epidermis; (3) it can transfect cells deep inside a tissue such as specific neurons within a brain slice; (4) it can accommodate mRNA, siRNA, or DNA practically of any size to be delivered; and (5) it works well with various cell types including non-dividing, terminally differentiated cells that are difficult to transfect, such as neurons or mammalian inner ear sensory hair cells. The latter advantage is particularly important for inner ear research. The disadvantages of this method are: (1) low efficiency of transfection due to many variables that have to be adjusted and (2) potential mechanical damage of the tissue if the biolistic shot parameters are not optimal. This chapter provides a step-by-step protocol and critical evaluation of the Bio-Rad Helios(®) Gene Gun transfection method used to deliver green fluorescent protein (GFP)-tagged full-length cDNAs of myosin 15a, whirlin, β-actin, and Clic5 into rodent hair cells of the postnatal inner ear sensory epithelia in culture.
Collapse
|