1
|
Siontas O, Ahn S. Challenges in AAV-Based Retinal Gene Therapies and the Role of Magnetic Nanoparticle Platforms. J Clin Med 2024; 13:7385. [PMID: 39685843 DOI: 10.3390/jcm13237385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Retinal diseases, leading to various visual impairments and blindness, are on the rise. However, the advancement of retinal gene therapies offers new hope for treatment of such diseases. Among different vector systems for conferring therapeutic genetic load to retinal cells, adeno-associated viruses (AAVs) have been most intensively explored and have already successfully gained multiple clinical approvals. AAV-based retinal gene therapies have shown great promise in treating retinal disorders, but usually rely on the heavily disruptive administration methods such as subretinal injection. This is because the clinically well-established, minimally invasive alternative of intravitreal injection (IVI) necessitates AAVs to traverse the retinal inner limiting membrane (ILM), which is hard to penetrate in higher eye models, like human or porcine eyes. Additionally, AAVs' natural transduction preference, known as tropism, is commonly not specific to cells of only one target retinal layer, which is another ongoing challenge in retinal gene therapy. This review examines strategies to overcome these obstacles with a focus on the potential of magnetic nanoparticles (MNPs) for improved retinal AAV delivery.
Collapse
Affiliation(s)
- Oliver Siontas
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4056 Basel, Switzerland
| | - Seungkuk Ahn
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4056 Basel, Switzerland
- UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
2
|
Ahn S, Siontas O, Koester J, Krol J, Fauser S, Müller DJ. Magnetically Guided Adeno-Associated Virus Delivery for the Spatially Targeted Transduction of Retina in Eyes. Adv Healthc Mater 2024; 13:e2401577. [PMID: 38848510 DOI: 10.1002/adhm.202401577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Indexed: 06/09/2024]
Abstract
Adeno-associated viruses (AAVs) are intensively explored for gene therapies in general and have found promising applications for treating retina diseases. However, controlling the specificity (tropism) and delivery of AAVs to selected layers, cell types, and areas of the retina is a major challenge to further develop retinal gene therapies. Magnetic nanoparticles (MNPs) provide effective delivery platforms to magnetically guide therapeutics to target cells. Yet, how MNPs can deliver AAVs to transfect particular retina layers and cells remains elusive. Here, it is demonstrated that MNPs can be used to transport different AAVs through the retina and to modulate the selective transduction of specific retinal layers or photoreceptor cells in ex vivo porcine explants and whole eyes. Thereby, transduction is triggered by bringing the viruses in close proximity to the target cell layer and by controlling their interaction time. It is shown that this magnetically guided approach to transport AAVs to selected areas and layers of the retina does not require the cell-specific optimization of the AAV tropism. It is anticipated that the new approach to control the delivery of AAVs and to selectively transduce cellular systems can be applied to many other tissues or organs to selectively deliver genes of interest.
Collapse
Affiliation(s)
- Seungkuk Ahn
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, Basel, 4056, Switzerland
| | - Oliver Siontas
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, Basel, 4056, Switzerland
| | - Janis Koester
- F. Hoffmann-La Roche Ltd, Roche Pharma Research and Early Development, Department of Ophthalmology, Basel, 4070, Switzerland
| | - Jacek Krol
- F. Hoffmann-La Roche Ltd, Roche Pharma Research and Early Development, Department of Ophthalmology, Basel, 4070, Switzerland
| | - Sascha Fauser
- F. Hoffmann-La Roche Ltd, Roche Pharma Research and Early Development, Department of Ophthalmology, Basel, 4070, Switzerland
| | - Daniel J Müller
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, Basel, 4056, Switzerland
| |
Collapse
|
3
|
Wei J, Liu C, Liang W, Yang X, Han S. Advances in optical molecular imaging for neural visualization. Front Bioeng Biotechnol 2023; 11:1250594. [PMID: 37671191 PMCID: PMC10475611 DOI: 10.3389/fbioe.2023.1250594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Iatrogenic nerve injury is a significant complication in surgery, which can negatively impact patients' quality of life. Currently, the main clinical neuroimaging methods, such as computed tomography, magnetic resonance imaging, and high-resolution ultrasonography, do not offer precise real-time positioning images for doctors during surgery. The clinical application of optical molecular imaging technology has led to the emergence of new concepts such as optical molecular imaging surgery, targeted surgery, and molecular-guided surgery. These advancements have made it possible to directly visualize surgical target areas, thereby providing a novel method for real-time identification of nerves during surgery planning. Unlike traditional white light imaging, optical molecular imaging technology enables precise positioning and identifies the cation of intraoperative nerves through the presentation of color images. Although a large number of experiments and data support its development, there are few reports on its actual clinical application. This paper summarizes the research results of optical molecular imaging technology and its ability to realize neural visualization. Additionally, it discusses the challenges neural visualization recognition faces and future development opportunities.
Collapse
Affiliation(s)
- Jinzheng Wei
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Chao Liu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenkai Liang
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaofeng Yang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shufeng Han
- Department of Orthopaedics, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
de Oliveira LF, Filho DM, Marques BL, Maciel GF, Parreira RC, do Carmo Neto JR, Da Silva PEF, Guerra RO, da Silva MV, Santiago HDC, Birbrair A, Kihara AH, Dias da Silva VJ, Glaser T, Resende RR, Ulrich H. Organoids as a novel tool in modelling infectious diseases. Semin Cell Dev Biol 2023; 144:87-96. [PMID: 36182613 DOI: 10.1016/j.semcdb.2022.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/04/2022] [Indexed: 11/23/2022]
Abstract
Infectious diseases worldwide affect human health and have important societal impacts. A better understanding of infectious diseases is urgently needed. In vitro and in vivo infection models have brought notable contributions to the current knowledge of these diseases. Organoids are multicellular culture systems resembling tissue architecture and function, recapitulating many characteristics of human disease and elucidating mechanisms of host-infectious agent interactions in the respiratory and gastrointestinal systems, the central nervous system and the skin. Here, we discuss the applicability of the organoid technology for modeling pathogenesis, host response and features, which can be explored for the development of preventive and therapeutic treatments.
Collapse
Affiliation(s)
- Lucas Felipe de Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Daniel Mendes Filho
- Departamento de Fisiologia, Escola Médica de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Bruno Lemes Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal deGoiás, Goiânia, GO, Brazil
| | | | | | - José Rodrigues do Carmo Neto
- Departamento de Biociência e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Rhanoica Oliveira Guerra
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Marcos Vinicius da Silva
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA; Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexandre H Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Valdo José Dias da Silva
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Talita Glaser
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil; Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Schubert R, Bae T, Simic B, Smith SN, Park SH, Nagy-Davidescu G, Gradinaru V, Plückthun A, Hur JK. CRISPR-clear imaging of melanin-rich B16-derived solid tumors. Commun Biol 2023; 6:370. [PMID: 37016073 PMCID: PMC10073193 DOI: 10.1038/s42003-023-04614-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/21/2023] [Indexed: 04/06/2023] Open
Abstract
Tissue clearing combined with deep imaging has emerged as a powerful technology to expand classical histological techniques. Current techniques have been optimized for imaging sparsely pigmented organs such as the mammalian brain. In contrast, melanin-rich pigmented tissue, of great interest in the investigation of melanomas, remains challenging. To address this challenge, we have developed a CRISPR-based gene editing approach that is easily incorporated into existing tissue-clearing workflows such the PACT clearing method. We term this method CRISPR-Clear. We demonstrate its applicability to highly melanin-rich B16-derived solid tumors, including one made transgenic for HER2, constituting one of very few syngeneic mouse tumors that can be used in immunocompetent models. We demonstrate the utility in detailed tumor characterization by staining for targeting antibodies and nanoparticles, as well as expressed fluorescent proteins. With CRISPR-Clear we have unprecedented access to optical interrogation in considerable portions of intact melanoma tissue for stained surface markers, expressed fluorescent proteins, of subcellular compartments, and of the vasculature.
Collapse
Affiliation(s)
- Rajib Schubert
- Department of Biochemistry, University of Zürich, Zürich, Switzerland.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Research and early development, Roche Sequencing Solutions, Pleasanton, CA, USA.
| | - Taegeun Bae
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
- College of Pharmacy, The Catholic University of Korea, Gyeonggi-do, South Korea
| | - Branko Simic
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
- Vector BioPharma AG, Basel, Switzerland
| | - Sheena N Smith
- Department of Biochemistry, University of Zürich, Zürich, Switzerland
- Vector BioPharma AG, Basel, Switzerland
| | - Seong-Ho Park
- Department of Medicine, Major in Medical Genetics, Graduate School, Hanyang University, Seoul, South Korea
| | | | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Zürich, Switzerland.
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, South Korea.
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
6
|
Mukherjee P, Park SH, Pathak N, Patino CA, Bao G, Espinosa HD. Integrating Micro and Nano Technologies for Cell Engineering and Analysis: Toward the Next Generation of Cell Therapy Workflows. ACS NANO 2022; 16:15653-15680. [PMID: 36154011 DOI: 10.1021/acsnano.2c05494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The emerging field of cell therapy offers the potential to treat and even cure a diverse array of diseases for which existing interventions are inadequate. Recent advances in micro and nanotechnology have added a multitude of single cell analysis methods to our research repertoire. At the same time, techniques have been developed for the precise engineering and manipulation of cells. Together, these methods have aided the understanding of disease pathophysiology, helped formulate corrective interventions at the cellular level, and expanded the spectrum of available cell therapeutic options. This review discusses how micro and nanotechnology have catalyzed the development of cell sorting, cellular engineering, and single cell analysis technologies, which have become essential workflow components in developing cell-based therapeutics. The review focuses on the technologies adopted in research studies and explores the opportunities and challenges in combining the various elements of cell engineering and single cell analysis into the next generation of integrated and automated platforms that can accelerate preclinical studies and translational research.
Collapse
Affiliation(s)
- Prithvijit Mukherjee
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - So Hyun Park
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Nibir Pathak
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| | - Cesar A Patino
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Gang Bao
- Department of Bioengineering, Rice University, 6500 Main Street, Houston, Texas 77030, United States
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Theoretical and Applied Mechanics Program, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
7
|
Abstract
The effect of the on-going COVID-19 pandemic on global healthcare systems has underlined the importance of timely and cost-effective point-of-care diagnosis of viruses. The need for ultrasensitive easy-to-use platforms has culminated in an increased interest for rapid response equipment-free alternatives to conventional diagnostic methods such as polymerase chain reaction, western-blot assay, etc. Furthermore, the poor stability and the bleaching behavior of several contemporary fluorescent reporters is a major obstacle in understanding the mechanism of viral infection thus retarding drug screening and development. Owing to their extraordinary surface-to-volume ratio as well as their quantum confinement and charge transfer properties, nanomaterials are desirable additives to sensing and imaging systems to amplify their signal response as well as temporal resolution. Their large surface area promotes biomolecular integration as well as efficacious signal transduction. Due to their hole mobility, photostability, resistance to photobleaching, and intense brightness, nanomaterials have a considerable edge over organic dyes for single virus tracking. This paper reviews the state-of-the-art of combining carbon-allotrope, inorganic and organic-based nanomaterials with virus sensing and tracking methods, starting with the impact of human pathogenic viruses on the society. We address how different nanomaterials can be used in various virus sensing platforms (e.g. lab-on-a-chip, paper, and smartphone-based point-of-care systems) as well as in virus tracking applications. We discuss the enormous potential for the use of nanomaterials as simple, versatile, and affordable tools for detecting and tracing viruses infectious to humans, animals, plants as well as bacteria. We present latest examples in this direction by emphasizing major advantages and limitations.
Collapse
Affiliation(s)
- Muqsit Pirzada
- Technical University of Berlin, Faculty of Natural Sciences and Maths, Straße des 17. Juni 124, Berlin 10623, Germany. .,Institute of Materials Science, Faculty of Engineering, Kiel University, Kaiserstr 2, 24143 Kiel, Germany
| | - Zeynep Altintas
- Technical University of Berlin, Faculty of Natural Sciences and Maths, Straße des 17. Juni 124, Berlin 10623, Germany. .,Institute of Materials Science, Faculty of Engineering, Kiel University, Kaiserstr 2, 24143 Kiel, Germany
| |
Collapse
|
8
|
Jacobsen RI, Nair RR, Obenhaus HA, Donato F, Slettmoen T, Moser MB, Moser EI. All-viral tracing of monosynaptic inputs to single birthdate-defined neurons in the intact brain. CELL REPORTS METHODS 2022; 2:100221. [PMID: 35637903 PMCID: PMC9142754 DOI: 10.1016/j.crmeth.2022.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/14/2022] [Accepted: 04/26/2022] [Indexed: 11/25/2022]
Abstract
Neuronal firing patterns are the result of inputs converging onto single cells. Identifying these inputs, anatomically and functionally, is essential to understand how neurons integrate information. Single-cell electroporation of helper genes and subsequent local injection of recombinant rabies viruses enable precise mapping of inputs to individual cells in superficial layers of the intact cortex. However, access to neurons in deeper structures requires more invasive procedures, including removal of overlying tissue. We developed a method that, through a combination of virus injections, allows us to target 4 or fewer hippocampal cells 48% of the time and a single cell 16% of the time in wild-type mice without use of electroporation or tissue aspiration. We identify local and distant monosynaptic inputs that can be functionally characterized in vivo. By expanding the toolbox for monosynaptic circuit tracing, this method will help further our understanding of neuronal integration at the level of single cells.
Collapse
Affiliation(s)
- R. Irene Jacobsen
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - Rajeevkumar R. Nair
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - Horst A. Obenhaus
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - Flavio Donato
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - Torstein Slettmoen
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - May-Britt Moser
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| | - Edvard I. Moser
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim 7030, Norway
| |
Collapse
|
9
|
Depla JA, Mulder LA, de Sá RV, Wartel M, Sridhar A, Evers MM, Wolthers KC, Pajkrt D. Human Brain Organoids as Models for Central Nervous System Viral Infection. Viruses 2022; 14:v14030634. [PMID: 35337041 PMCID: PMC8948955 DOI: 10.3390/v14030634] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Pathogenesis of viral infections of the central nervous system (CNS) is poorly understood, and this is partly due to the limitations of currently used preclinical models. Brain organoid models can overcome some of these limitations, as they are generated from human derived stem cells, differentiated in three dimensions (3D), and can mimic human neurodevelopmental characteristics. Therefore, brain organoids have been increasingly used as brain models in research on various viruses, such as Zika virus, severe acute respiratory syndrome coronavirus 2, human cytomegalovirus, and herpes simplex virus. Brain organoids allow for the study of viral tropism, the effect of infection on organoid function, size, and cytoarchitecture, as well as innate immune response; therefore, they provide valuable insight into the pathogenesis of neurotropic viral infections and testing of antivirals in a physiological model. In this review, we summarize the results of studies on viral CNS infection in brain organoids, and we demonstrate the broad application and benefits of using a human 3D model in virology research. At the same time, we describe the limitations of the studies in brain organoids, such as the heterogeneity in organoid generation protocols and age at infection, which result in differences in results between studies, as well as the lack of microglia and a blood brain barrier.
Collapse
Affiliation(s)
- Josse A. Depla
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC Location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.A.M.); (A.S.); (K.C.W.); (D.P.)
- Department of Pediatric Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- UniQure Biopharma B.V., Department of Research & Development, Paasheuvelweg 25A, 1105 BE Amsterdam, The Netherlands; (R.V.d.S.); (M.W.); (M.M.E.)
- Correspondence:
| | - Lance A. Mulder
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC Location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.A.M.); (A.S.); (K.C.W.); (D.P.)
- Department of Pediatric Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Renata Vieira de Sá
- UniQure Biopharma B.V., Department of Research & Development, Paasheuvelweg 25A, 1105 BE Amsterdam, The Netherlands; (R.V.d.S.); (M.W.); (M.M.E.)
| | - Morgane Wartel
- UniQure Biopharma B.V., Department of Research & Development, Paasheuvelweg 25A, 1105 BE Amsterdam, The Netherlands; (R.V.d.S.); (M.W.); (M.M.E.)
| | - Adithya Sridhar
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC Location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.A.M.); (A.S.); (K.C.W.); (D.P.)
| | - Melvin M. Evers
- UniQure Biopharma B.V., Department of Research & Development, Paasheuvelweg 25A, 1105 BE Amsterdam, The Netherlands; (R.V.d.S.); (M.W.); (M.M.E.)
| | - Katja C. Wolthers
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC Location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.A.M.); (A.S.); (K.C.W.); (D.P.)
| | - Dasja Pajkrt
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC Location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (L.A.M.); (A.S.); (K.C.W.); (D.P.)
- Department of Pediatric Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
10
|
Lehtinen K, Nokia MS, Takala H. Red Light Optogenetics in Neuroscience. Front Cell Neurosci 2022; 15:778900. [PMID: 35046775 PMCID: PMC8761848 DOI: 10.3389/fncel.2021.778900] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/02/2021] [Indexed: 12/25/2022] Open
Abstract
Optogenetics, a field concentrating on controlling cellular functions by means of light-activated proteins, has shown tremendous potential in neuroscience. It possesses superior spatiotemporal resolution compared to the surgical, electrical, and pharmacological methods traditionally used in studying brain function. A multitude of optogenetic tools for neuroscience have been created that, for example, enable the control of action potential generation via light-activated ion channels. Other optogenetic proteins have been used in the brain, for example, to control long-term potentiation or to ablate specific subtypes of neurons. In in vivo applications, however, the majority of optogenetic tools are operated with blue, green, or yellow light, which all have limited penetration in biological tissues compared to red light and especially infrared light. This difference is significant, especially considering the size of the rodent brain, a major research model in neuroscience. Our review will focus on the utilization of red light-operated optogenetic tools in neuroscience. We first outline the advantages of red light for in vivo studies. Then we provide a brief overview of the red light-activated optogenetic proteins and systems with a focus on new developments in the field. Finally, we will highlight different tools and applications, which further facilitate the use of red light optogenetics in neuroscience.
Collapse
Affiliation(s)
- Kimmo Lehtinen
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Miriam S. Nokia
- Department of Psychology, University of Jyväskylä, Jyväskylä, Finland
- Centre for Interdisciplinary Brain Research, University of Jyväskylä, Jyväskylä, Finland
| | - Heikki Takala
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
11
|
AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat Neurosci 2022; 25:106-115. [PMID: 34887588 DOI: 10.1038/s41593-021-00969-4] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/22/2021] [Indexed: 11/09/2022]
Abstract
Genetic intervention is increasingly being explored as a therapeutic option for debilitating disorders of the central nervous system. The safety and efficacy of gene therapies rely upon expressing a transgene in affected cells while minimizing off-target expression. Here we show organ-specific targeting of adeno-associated virus (AAV) capsids after intravenous delivery, which we achieved by employing a Cre-transgenic-based screening platform and sequential engineering of AAV-PHP.eB between the surface-exposed AA452 and AA460 of VP3. From this selection, we identified capsid variants that were enriched in the brain and targeted away from the liver in C57BL/6J mice. This tropism extends to marmoset (Callithrix jacchus), enabling robust, non-invasive gene delivery to the marmoset brain after intravenous administration. Notably, the capsids identified result in distinct transgene expression profiles within the brain, with one exhibiting high specificity to neurons. The ability to cross the blood-brain barrier with neuronal specificity in rodents and non-human primates enables new avenues for basic research and therapeutic possibilities unattainable with naturally occurring serotypes.
Collapse
|
12
|
Magnetically Single-Cell Virus Stamping. Methods Mol Biol 2021. [PMID: 34228300 DOI: 10.1007/978-1-0716-1441-9_20] [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
Single-cell engineering via virus based genetic manipulation allows the possibility of understanding of complex tissues. However, current delivery methods for the genetic engineering of single cells via viral transduction suffer from limitations that restrict their application. Here I present a protocol describing a precise technique which can be used for the targeted virus infection of single cells in a monolayer of cells that is optically accessible. The protocol, demonstrated here by stamping cultured Hela cells with lentiviruses (LVs), completes in a few minutes and allows stable transgene expression within a few days, at success rates approaching 80%.
Collapse
|
13
|
Völkner M, Pavlou M, Büning H, Michalakis S, Karl MO. Optimized Adeno-Associated Virus Vectors for Efficient Transduction of Human Retinal Organoids. Hum Gene Ther 2021; 32:694-706. [PMID: 33752467 DOI: 10.1089/hum.2020.321] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The most widely used vectors for gene delivery in the retina are recombinant adeno-associated virus (rAAV) vectors. They have proven to be safe and effective in retinal gene therapy studies aimed to treat inherited retinal dystrophies, although with various limitations in transduction efficiency. Novel variants with modified capsid sequences have been engineered to improve transduction and overcome limitations of naturally occurring variants. Although preclinical evaluation of rAAV vectors based on such novel capsids is mostly done in animal models, the use of human induced pluripotent stem cell (hiPSC)-derived organoids offers an accessible and abundant human testing platform for rAAV evaluation. In this study, we tested the novel capsids, AAV9.GL and AAV9.NN, for their tropism and transduction efficiency in hiPSC-derived human retinal organoids (HROs) with all major neuronal and glial cell types in a laminated structure. These variants are based on the AAV9 capsid and were engineered to display specific surface-exposed peptide sequences, previously shown to improve the retinal transduction properties in the context of AAV2. To this end, HROs were transduced with increasing concentrations of rAAV9, rAAV9.GL, or rAAV9.NN carrying a self-complementary genome with a cytomegalovirus-enhanced green fluorescent protein (eGFP) cassette and were monitored for eGFP expression. The rAAV vectors transduced HROs in a dose-dependent manner, with rAAV9.NN achieving the highest efficiency and fastest onset kinetics, leading to detectable eGFP signals in photoreceptors, some interneurons, and Müller glia already at 2 days post-transduction. The potency-enhancing effect of the NN peptide insert was replicated when using the corresponding AAV2-based version (rAAV2.NN). Taken together, we report the application of an HRO system for screening novel AAV vectors and introduce novel vector candidates with enhanced transduction efficiency for human retinal cells.
Collapse
Affiliation(s)
- Manuela Völkner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Marina Pavlou
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany.,Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany.,Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mike O Karl
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.,CRTD-Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.,TU Dresden, Faculty of Medicine Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
14
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
15
|
Xu YP, Zhou HY, Wang GC, Zhang Y, Yang T, Zhao Y, Li RT, Zhang RR, Guo Y, Wang X, Li XF, Qin CF, Tang R. Rational Design of a Replication-Competent and Inheritable Magnetic Viruses for Targeting Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002435. [PMID: 32954651 DOI: 10.1002/smll.202002435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/20/2020] [Indexed: 06/11/2023]
Abstract
Infection with live-attenuated vaccines always inevitably induces side effects that reduce their safety. This study suggests a concept of magnetic virus produced by genetically modifying viral surfaces with Fe3 O4 nanoparticles (NPs) to control their tropisms. An iron-affinity peptide is designed to be displayed on the viral surface protein (VP1) of human enterovirus type 71 (EV71), a typical nonenveloped picornavirus, as the model. The modified EV71 can self-bind with Fe3 O4 NPs under physiological conditions, resulting in novel EV71-Fe3 O4 hybrid materials. This rationally engineered EV71 with Fe3 O4 retains its original biological infectivity, but its tropism can be precisely controlled by magnetism. Both in vitro and in vivo experiments demonstrate that EV71-Fe3 O4 can infect only a desired area within the limit of the applied magnetic field, which effectively reduces its pathological damage. More importantly, this characteristic of EV71 can be inherited due to the gene-induced coassembly of viruses and NPs. This achievement provides a proof of concept in virus vaccine improvement by a combination of gene modification and material incorporation, leading to great potential for biomedical developments.
Collapse
Affiliation(s)
- Yan-Peng Xu
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hang-Yu Zhou
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
- Suzhou Institute of System Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215000, China
| | - Guang-Chuan Wang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Ying Zhang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Tianxu Yang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Yueqi Zhao
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Rui-Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xiaoyu Wang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Ruikang Tang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| |
Collapse
|
16
|
Rangel Olguin AG, Rochon PL, Krishnaswamy A. New Optical Tools to Study Neural Circuit Assembly in the Retina. Front Neural Circuits 2020; 14:44. [PMID: 32848633 PMCID: PMC7424070 DOI: 10.3389/fncir.2020.00044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
During development, neurons navigate a tangled thicket of thousands of axons and dendrites to synapse with just a few specific targets. This phenomenon termed wiring specificity, is critical to the assembly of neural circuits and the way neurons manage this feat is only now becoming clear. Recent studies in the mouse retina are shedding new insight into this process. They show that specific wiring arises through a series of stages that include: directed axonal and dendritic growth, the formation of neuropil layers, positioning of such layers, and matching of co-laminar synaptic partners. Each stage appears to be directed by a distinct family of recognition molecules, suggesting that the combinatorial expression of such family members might act as a blueprint for retinal connectivity. By reviewing the evidence in support of each stage, and by considering their underlying molecular mechanisms, we attempt to synthesize these results into a wiring model which generates testable predictions for future studies. Finally, we conclude by highlighting new optical methods that could be used to address such predictions and gain further insight into this fundamental process.
Collapse
|
17
|
Tay A. The Benefits of Going Small: Nanostructures for Mammalian Cell Transfection. ACS NANO 2020; 14:7714-7721. [PMID: 32631053 DOI: 10.1021/acsnano.0c04624] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanostructures, with their localized interactions with mammalian cells, can offer better efficiency and lower cell perturbation than conventional viral, biochemical, and electroporation transfection techniques. In this Perspective, I describe the different stages of transfection and provide a comparison of transfection techniques based on their mechanisms. Focusing on specific aims of transfection, I also illustrate how recent developments in high-aspect-ratio nanostructures have endowed them with properties that are superior to existing viral, biochemical, and electroporation methods as a versatile technique to deliver a variety of cargoes and to interface with different mammalian cell types for biomedical applications. Finally, I describe the challenges associated with transfection that need to be overcome to enhance cargo delivery efficiency and clinical translation.
Collapse
Affiliation(s)
- Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| |
Collapse
|
18
|
Batabyal S, Gajjeraman S, Tchedre K, Dibas A, Wright W, Mohanty S. Near-Infrared Laser-Based Spatially Targeted Nano-enhanced Optical Delivery of Therapeutic Genes to Degenerated Retina. Mol Ther Methods Clin Dev 2020; 17:758-770. [PMID: 32355865 PMCID: PMC7184107 DOI: 10.1016/j.omtm.2020.03.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022]
Abstract
Non-viral delivery of therapeutic genes into targeted areas of retina is essential for re-functionalizing the retinal circuitry. While a focused ultrafast laser beam has been recently used for intra-ocular delivery of molecules, it poses the significant technical challenge of overcoming aberrations of the eye and maintaining a tightly focused spot on the retinal cell membrane. Furthermore, to minimize collateral damage and increase the throughput of gene delivery, we introduced a weakly focused near-infrared (NIR) continuous wave (CW) or pulsed laser beam on to the cells wherein the intensity is locally enhanced by gold nanorods bound to the cell membranes to permit gene insertion. Parametric optimization of nano-enhanced optical delivery (NOD) was carried out by varying the exposure time, as well as the power of the CW NIR beam or the energy of the pulsed NIR beam. Using this NOD method, therapeutic genes encoding for multi-characteristic opsins (MCOs) were delivered to spatially targeted regions of degenerated retina ex vivo as well as in vivo. NOD-mediated cell membrane-specific expression of MCOs in targeted retinal regions with photoreceptor degeneration will allow functional recovery in an ambient light environment.
Collapse
Affiliation(s)
- Subrata Batabyal
- Nanoscope Technologies, 1312 Brown Trail, Bedford, TX 76022, USA
| | | | - Kissaou Tchedre
- Nanoscope Technologies, 1312 Brown Trail, Bedford, TX 76022, USA
| | - Adnan Dibas
- Nanoscope Technologies, 1312 Brown Trail, Bedford, TX 76022, USA
| | - Weldon Wright
- Nanoscope Technologies, 1312 Brown Trail, Bedford, TX 76022, USA
| | | |
Collapse
|
19
|
Kohara K, Inoue A, Nakano Y, Hirai H, Kobayashi T, Maruyama M, Baba R, Kawashima C. BATTLE: Genetically Engineered Strategies for Split-Tunable Allocation of Multiple Transgenes in the Nervous System. iScience 2020; 23:101248. [PMID: 32629613 PMCID: PMC7322263 DOI: 10.1016/j.isci.2020.101248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/02/2019] [Accepted: 06/04/2020] [Indexed: 11/26/2022] Open
Abstract
Elucidating fine architectures and functions of cellular and synaptic connections requires development of new flexible methods. Here, we created a concept called the “battle of transgenes,” based on which we generated strategies using genetically engineered battles of multiple recombinases. The strategies enabled split-tunable allocation of multiple transgenes. We demonstrated the versatility of these strategies and technologies in inducing strong and multi-sparse allocations of multiple transgenes. Furthermore, the combination of our transgenic strategy and expansion microscopy enabled three-dimensional high-resolution imaging of whole synaptic structures in the hippocampus with simultaneous visualizations of endogenous synaptic proteins. These strategies and technologies based on the battle of genes may accelerate the analysis of whole synaptic and cellular connections in diverse life science fields. Generation of BATTLE-recombinase systems for allocation of multiple transgenes Split-tunable allocation in BATTLE-1 and multi-sparse allocation in BATTLE-2 Clear and strong labeling of dendrites and axons using BATTLE-2 3D high-resolution imaging of whole synapses in hippocampus in BATTLE-1EX
Collapse
Affiliation(s)
- Keigo Kohara
- Department of Cellular and Functional Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan.
| | - Akitoshi Inoue
- Department of Medical Chemistry, Kansai Medical University, Graduate School of Medicine, Hirakata, Osaka 573-1010, Japan
| | - Yousuke Nakano
- Department of Anatomy, Kansai Medical University, Graduate School of Medicine, Hirakata, Osaka 573-1010, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; Research Program for Neural Signalling, Division of Endocrinology, Metabolism and Signal Research, Gunma University Initiative for Advanced Research, Maebashi, Gunma 371-8512, Japan
| | - Takuya Kobayashi
- Department of Medical Chemistry, Kansai Medical University, Graduate School of Medicine, Hirakata, Osaka 573-1010, Japan; Japan Agency for Medical Research and Development (AMED), Core Research for Evolutional Science and Technology (CREST), 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Masato Maruyama
- Department of Anatomy, Kansai Medical University, Graduate School of Medicine, Hirakata, Osaka 573-1010, Japan; Faculty of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Ryosuke Baba
- Department of Cellular and Functional Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Chiho Kawashima
- Department of Cellular and Functional Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan; Department of Bioscience, Osaka College of High Technology, Osaka 532-003, Japan
| |
Collapse
|
20
|
Brückner S, Schubert R, Kraushaar T, Hartmann R, Hoffmann D, Jelli E, Drescher K, Müller DJ, Oliver Essen L, Mösch HU. Kin discrimination in social yeast is mediated by cell surface receptors of the Flo11 adhesin family. eLife 2020; 9:55587. [PMID: 32286952 PMCID: PMC7156268 DOI: 10.7554/elife.55587] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Microorganisms have evolved specific cell surface molecules that enable discrimination between cells from the same and from a different kind. Here, we investigate the role of Flo11-type cell surface adhesins from social yeasts in kin discrimination. We measure the adhesion forces mediated by Flo11A-type domains using single-cell force spectroscopy, quantify Flo11A-based cell aggregation in populations and determine the Flo11A-dependent segregation of competing yeast strains in biofilms. We find that Flo11A domains from diverse yeast species confer remarkably strong adhesion forces by establishing homotypic interactions between single cells, leading to efficient cell aggregation and biofilm formation in homogenous populations. Heterotypic interactions between Flo11A domains from different yeast species or Saccharomyces cerevisiae strains confer weak adhesive forces and lead to efficient strain segregation in heterogenous populations, indicating that in social yeasts Flo11A-mediated cell adhesion is a major mechanism for kin discrimination at species and sub-species levels. These findings, together with our structure and mutation analysis of selected Flo11A domains, provide a rationale of how cell surface receptors have evolved in microorganisms to mediate kin discrimination.
Collapse
Affiliation(s)
- Stefan Brückner
- Department of Genetics, Philipps-Universität Marburg, Marburg, Germany
| | - Rajib Schubert
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Timo Kraushaar
- Department of Biochemistry, Philipps-Universität Marburg, Marburg, Germany
| | - Raimo Hartmann
- Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Daniel Hoffmann
- Department of Genetics, Philipps-Universität Marburg, Marburg, Germany
| | - Eric Jelli
- Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Knut Drescher
- Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Lars Oliver Essen
- Department of Biochemistry, Philipps-Universität Marburg, Marburg, Germany.,LOEWE Center for Synthetic Microbiology, Philipps-Universität Marburg, Marburg, Germany
| | - Hans-Ulrich Mösch
- Department of Genetics, Philipps-Universität Marburg, Marburg, Germany.,LOEWE Center for Synthetic Microbiology, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
21
|
Xiao M, Shen Z, Luo W, Tan B, Meng X, Wu X, Wu S, Nie K, Tong T, Hong J, Wang X, Wang X. A new colitis therapy strategy via the target colonization of magnetic nanoparticle-internalized Roseburia intestinalis. Biomater Sci 2020; 7:4174-4185. [PMID: 31380882 DOI: 10.1039/c9bm00980a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The homeostasis process in the gut tissue of humans relies on intestinal bacteria. However, the intestine is a complex structural tissue with a huge superficial area, and thus the effective application of probiotics in the treatment of Crohn's disease (CD) is still challenging. Herein, we show the feasibility of probiotic target delivery and retention using magnetic iron oxide nanoparticle-internalized Roseburia intestinalis, which can be easily directed by a magnetic field in vitro and in vivo. Subsequently, the increased colonization of this core profitable flora not only resulted in a better therapy effect than traditional intragastric administration but also altered the bacterial composition, leading to a higher diversity in microbial taxa in rats with colitis. Our findings illustrate the exciting opportunities that nanotechnology offers for alternative strategies to modulate biological systems remotely and precisely, which represent a step towards the wireless magnetic manipulation of living biological entities in microbiology.
Collapse
Affiliation(s)
- Mengwei Xiao
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Traber GL, Della Volpe-Waizel M, Maloca P, Schmidt-Erfurth U, Rubin G, Roska B, Cordeiro MF, Otto T, Weleber R, Lesmes LA, Arleo A, Scholl HPN. New Technologies for Outcome Measures in Glaucoma: Review by the European Vision Institute Special Interest Focus Group. Ophthalmic Res 2020; 63:88-96. [PMID: 31935739 DOI: 10.1159/000504892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/19/2019] [Indexed: 11/19/2022]
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide, with an increasing prevalence. The complexity of the disease has been a major challenge in moving the field forward with regard to both pathophysiological insight and treatment. In this context, discussing possible outcome measures in glaucoma trials is of utmost importance and clinical relevance. A recent meeting of the European Vision Institute (EVI) special interest focus group was held on "New Technologies for Outcome Measures in Retina and Glaucoma," addressing both functional and structural outcomes, as well as translational hot topics in glaucoma and retina research. In conjunction with the published literature, this review summarizes the meeting focusing on glaucoma.
Collapse
Affiliation(s)
- Ghislaine L Traber
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - Maria Della Volpe-Waizel
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - Peter Maloca
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | | | - Gary Rubin
- Institute of Ophthalmology, UCL University College London, London, United Kingdom
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - M Francesca Cordeiro
- Institute of Ophthalmology, UCL University College London, London, United Kingdom.,Western Eye Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.,Imperial College Ophthalmic Research Group (ICORG), Imperial College London, London, United Kingdom
| | - Tilman Otto
- Heidelberg Engineering GmbH, Heidelberg, Germany
| | - Richard Weleber
- Casey Eye Institute, Departments of Ophthalmology and Molecular and Medical Genetics, University of Oregon Health & Science University, Portland, Oregon, USA
| | | | - Angelo Arleo
- Institut de la Vision, CNRS, INSERM, Sorbonne Université, Paris, France
| | - Hendrik P N Scholl
- Department of Ophthalmology, University Hospital Basel, University of Basel, Basel, Switzerland, .,Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland,
| |
Collapse
|
23
|
Fischer J, Heide M, Huttner WB. Genetic Modification of Brain Organoids. Front Cell Neurosci 2019; 13:558. [PMID: 31920558 PMCID: PMC6928125 DOI: 10.3389/fncel.2019.00558] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
Brain organoids have become increasingly used systems allowing 3D-modeling of human brain development, evolution, and disease. To be able to make full use of these modeling systems, researchers have developed a growing toolkit of genetic modification techniques. These techniques can be applied to mature brain organoids or to the preceding embryoid bodies (EBs) and founding cells. This review will describe techniques used for transient and stable genetic modification of brain organoids and discuss their current use and respective advantages and disadvantages. Transient approaches include adeno-associated virus (AAV) and electroporation-based techniques, whereas stable genetic modification approaches make use of lentivirus (including viral stamping), transposon and CRISPR/Cas9 systems. Finally, an outlook as to likely future developments and applications regarding genetic modifications of brain organoids will be presented.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
24
|
Schubert R, Herzog S, Trenholm S, Roska B, Müller DJ. Magnetically guided virus stamping for the targeted infection of single cells or groups of cells. Nat Protoc 2019; 14:3205-3219. [DOI: 10.1038/s41596-019-0221-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 07/02/2019] [Indexed: 01/10/2023]
|
25
|
Frank JA, Antonini MJ, Anikeeva P. Next-generation interfaces for studying neural function. Nat Biotechnol 2019; 37:1013-1023. [PMID: 31406326 PMCID: PMC7243676 DOI: 10.1038/s41587-019-0198-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 06/26/2019] [Indexed: 01/06/2023]
Abstract
Monitoring and modulating the diversity of signals used by neurons and glia in a closed-loop fashion is necessary to establish causative links between biochemical processes within the nervous system and observed behaviors. As developments in neural-interface hardware strive to keep pace with rapid progress in genetically encoded and synthetic reporters and modulators of neural activity, the integration of multiple functional features becomes a key requirement and a pressing challenge in the field of neural engineering. Electrical, optical and chemical approaches have been used to manipulate and record neuronal activity in vivo, with a recent focus on technologies that both integrate multiple modes of interaction with neurons into a single device and enable bidirectional communication with neural circuits with enhanced spatiotemporal precision. These technologies not only are facilitating a greater understanding of the brain, spinal cord and peripheral circuits in the context of health and disease, but also are informing the development of future closed-loop therapies for neurological, neuro-immune and neuroendocrine conditions.
Collapse
Affiliation(s)
- James A Frank
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc-Joseph Antonini
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Science & Technology Graduate Program, Cambridge, MA, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Material Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
26
|
Schwarz MK, Remy S. Rabies virus-mediated connectivity tracing from single neurons. J Neurosci Methods 2019; 325:108365. [DOI: 10.1016/j.jneumeth.2019.108365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 02/01/2023]
|
27
|
Molecular Cloning, Lentiviral Transduction, and Expression of Recombinant ADAMTSL2 and ADAMTSL4. Methods Mol Biol 2019. [PMID: 31463909 DOI: 10.1007/978-1-4939-9698-8_12] [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
Lentiviral systems have proven advantageous in the delivery and long-term integration of gene sequences into the genome of several cell types in vitro, in vivo, as well as in clinical trials. Here we detail the protocols involved in the molecular cloning of ADAMTSL2 and ADAMTSL4 into the human immunodeficiency virus (HIV)-derived pCDH lentiviral system. We also describe the lentiviral transduction of ADAMTSL2 and ADAMTSL4 into mammalian HEK293-EBNA cells to create stable cell lines, as well as their recombinant expression.
Collapse
|
28
|
Obien MEJ, Frey U. Large-Scale, High-Resolution Microelectrode Arrays for Interrogation of Neurons and Networks. ADVANCES IN NEUROBIOLOGY 2019; 22:83-123. [PMID: 31073933 DOI: 10.1007/978-3-030-11135-9_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
High-density microelectrode arrays (HD-MEAs) are increasingly being used for the observation and manipulation of neurons and networks in vitro. Large-scale electrode arrays allow for long-term extracellular recording of the electrical activity from thousands of neurons simultaneously. Beyond population activity, it has also become possible to extract information of single neurons at subcellular level (e.g., the propagation of action potentials along axons). In effect, HD-MEAs have become an electrical imaging platform for label-free extraction of the structure and activation of cells in cultures and tissues. The quality of HD-MEA data depends on the resolution of the electrode array and the signal-to-noise ratio. In this chapter, we begin with an introduction to HD-MEA signals. We provide an overview of the developments on complementary metal-oxide-semiconductor or CMOS-based HD-MEA technology. We also discuss the factors affecting the performance of HD-MEAs and the trending application requirements that drive the efforts for future devices. We conclude with an outlook on the potential of HD-MEAs for advancing basic neuroscience and drug discovery.
Collapse
Affiliation(s)
- Marie Engelene J Obien
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
- MaxWell Biosystems, Basel, Switzerland.
| | - Urs Frey
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- MaxWell Biosystems, Basel, Switzerland
| |
Collapse
|
29
|
Della Volpe-Waizel M, Traber GL, Maloca P, Zinkernagel M, Schmidt-Erfurth U, Rubin G, Roska B, Otto T, Weleber RG, Scholl HPN. New Technologies for Outcome Measures in Retinal Disease: Review from the European Vision Institute Special Interest Focus Group. Ophthalmic Res 2019; 63:77-87. [PMID: 31352462 DOI: 10.1159/000501887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/03/2019] [Indexed: 01/03/2023]
Abstract
Novel diagnostic tools to measure retinal function and structure are rapidly being developed and introduced into clinical use. Opportunities exist to use these informative and robust measures as endpoints for clinical trials to determine efficacy and to monitor safety of therapeutic interventions. In order to inform researchers and clinician-scientists about these new diagnostic tools, a workshop was organized by the European Vision Institute. Invited speakers highlighted the recent advances in state-of-the-art technologies for outcome measures in the field of retina. This review highlights the workshop's presentations in the context of published literature.
Collapse
Affiliation(s)
- Maria Della Volpe-Waizel
- Department of Ophthalmology, University of Basel, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Ghislaine L Traber
- Department of Ophthalmology, University of Basel, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Peter Maloca
- Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Martin Zinkernagel
- Department of Ophthalmology and Department of Clinical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Gary Rubin
- UCL University College London, Institute of Ophthalmology, London, United Kingdom
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Tilman Otto
- Heidelberg Engineering GmbH, Heidelberg, Germany
| | - Richard G Weleber
- Casey Eye Institute, Departments of Ophthalmology and Molecular and Medical Genetics, University of Oregon Health and Science University, Portland, Oregon, USA
| | - Hendrik P N Scholl
- Department of Ophthalmology, University of Basel, Basel, Switzerland, .,Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland,
| |
Collapse
|
30
|
Wan Z, Shaheen S, Chau A, Zeng Y, Liu W. Imaging: Gear up for mechano-immunology. Cell Immunol 2019; 350:103926. [PMID: 31151736 DOI: 10.1016/j.cellimm.2019.103926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 04/15/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Immune cells including B and T lymphocytes have a remarkable ability to sense the physical perturbations through their surface expressed receptors. At the advent of modern imaging technologies paired with biophysical methods, we have gained the understanding of mechanical forces exerted by immune cells to perform their functions. This review will go over the imaging techniques already being used to study mechanical forces in immune cells. We will also discuss the dire need for new modern technologies for future work.
Collapse
Affiliation(s)
- Zhengpeng Wan
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Samina Shaheen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Alicia Chau
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
31
|
Kolb I, Landry CR, Yip MC, Lewallen CF, Stoy WA, Lee J, Felouzis A, Yang B, Boyden ES, Rozell CJ, Forest CR. PatcherBot: a single-cell electrophysiology robot for adherent cells and brain slices. J Neural Eng 2019; 16:046003. [PMID: 30970335 DOI: 10.1088/1741-2552/ab1834] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Intracellular patch-clamp electrophysiology, one of the most ubiquitous, high-fidelity techniques in biophysics, remains laborious and low-throughput. While previous efforts have succeeded at automating some steps of the technique, here we demonstrate a robotic 'PatcherBot' system that can perform many patch-clamp recordings sequentially, fully unattended. APPROACH Comprehensive automation is accomplished by outfitting the robot with machine vision, and cleaning pipettes instead of manually exchanging them. MAIN RESULTS the PatcherBot can obtain data at a rate of 16 cells per hour and work with no human intervention for up to 3 h. We demonstrate the broad applicability and scalability of this system by performing hundreds of recordings in tissue culture cells and mouse brain slices with no human supervision. Using the PatcherBot, we also discovered that pipette cleaning can be improved by a factor of three. SIGNIFICANCE The system is potentially transformative for applications that depend on many high-quality measurements of single cells, such as drug screening, protein functional characterization, and multimodal cell type investigations.
Collapse
Affiliation(s)
- Ilya Kolb
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States of America
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Roska B. The first steps in vision: cell types, circuits, and repair. EMBO Mol Med 2019; 11:e10218. [PMID: 30670464 PMCID: PMC6404109 DOI: 10.15252/emmm.201810218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 11/09/2022] Open
Abstract
Dysfunction of the key sense of vision, leading to visual handicap or blindness, has a crucial effect on day-to-day life. In this commentary, I will summarize the work in my laboratory that is focused on a basic understanding of visual processing and the use of this information to understand disease mechanism and to develop correcting therapies. We are beginning to understand how cell types of the visual system interact in local circuits and compute visual information. This has brought insight into mechanisms of cell-type-specific diseases and has allowed us to design new therapies for restoring vision in genetic forms of blindness.
Collapse
Affiliation(s)
- Botond Roska
- Institute of Molecular and Clinical Ophthalmology BaselBaselSwitzerland
- University of BaselBaselSwitzerland
- Friedrich Miescher InstituteBaselSwitzerland
| |
Collapse
|
33
|
Obien MEJ, Hierlemann A, Frey U. Accurate signal-source localization in brain slices by means of high-density microelectrode arrays. Sci Rep 2019; 9:788. [PMID: 30692552 PMCID: PMC6349853 DOI: 10.1038/s41598-018-36895-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/28/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular recordings by means of high-density microelectrode arrays (HD-MEAs) have become a powerful tool to resolve subcellular details of single neurons in active networks grown from dissociated cells. To extend the application of this technology to slice preparations, we developed models describing how extracellular signals, produced by neuronal cells in slices, are detected by microelectrode arrays. The models help to analyze and understand the electrical-potential landscape in an in vitro HD-MEA-recording scenario based on point-current sources. We employed two modeling schemes, (i) a simple analytical approach, based on the method of images (MoI), and (ii) an approach, based on finite-element methods (FEM). We compared and validated the models with large-scale, high-spatiotemporal-resolution recordings of slice preparations by means of HD-MEAs. We then developed a model-based localization algorithm and compared the performance of MoI and FEM models. Both models provided accurate localization results and a comparable and negligible systematic error, when the point source was in saline, a condition similar to cell-culture experiments. Moreover, the relative random error in the x-y-z-localization amounted only up to 4.3% for z-distances up to 200 μm from the HD-MEA surface. In tissue, the systematic errors of both, MoI and FEM models were significantly higher, and a pre-calibration was required. Nevertheless, the FEM values proved to be closer to the tissue experimental results, yielding 5.2 μm systematic mean error, compared to 22.0 μm obtained with MoI. These results suggest that the medium volume or "saline height", the brain slice thickness and anisotropy, and the location of the reference electrode, which were included in the FEM model, considerably affect the extracellular signal and localization performance, when the signal source is at larger distance to the array. After pre-calibration, the relative random error of the z-localization in tissue was only 3% for z-distances up to 200 μm. We then applied the model and related detailed understanding of extracellular recordings to achieve an electrically-guided navigation of a stimulating micropipette, solely based on the measured HD-MEA signals, and managed to target spontaneously active neurons in an acute brain slice for electroporation.
Collapse
Affiliation(s)
- Marie Engelene J Obien
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- RIKEN Quantitative Biology Center, Kobe, Japan.
- MaxWell Biosystems AG, Basel, Switzerland.
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Urs Frey
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- RIKEN Quantitative Biology Center, Kobe, Japan
- MaxWell Biosystems AG, Basel, Switzerland
| |
Collapse
|
34
|
Lin R, Wang R, Yuan J, Feng Q, Zhou Y, Zeng S, Ren M, Jiang S, Ni H, Zhou C, Gong H, Luo M. Cell-type-specific and projection-specific brain-wide reconstruction of single neurons. Nat Methods 2018; 15:1033-1036. [PMID: 30455464 DOI: 10.1038/s41592-018-0184-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/31/2018] [Indexed: 11/09/2022]
Abstract
We developed a dual-adeno-associated-virus expression system that enables strong and sparse labeling of individual neurons with cell-type and projection specificity. We demonstrated its utility for whole-brain reconstruction of midbrain dopamine neurons and striatum-projecting cortical neurons. We further extended the labeling method for rapid reconstruction in cleared thick brain sections and simultaneous dual-color labeling. This labeling system may facilitate the process of generating mesoscale single-neuron projectomes of mammalian brains.
Collapse
Affiliation(s)
- Rui Lin
- School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Ruiyu Wang
- School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Qiru Feng
- National Institute of Biological Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Youtong Zhou
- National Institute of Biological Sciences, Beijing, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Ren
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Jiang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ni
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Can Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China. .,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China. .,HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China.
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China. .,School of Life Sciences, Tsinghua University, Beijing, China. .,Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
35
|
Zhang T, Huang Y, Ma X, Gong N, Liu X, Liu L, Ye X, Hu B, Li C, Tian JH, Magrini A, Zhang J, Guo W, Xing JF, Bottini M, Liang XJ. Fluorinated Oligoethylenimine Nanoassemblies for Efficient siRNA-Mediated Gene Silencing in Serum-Containing Media by Effective Endosomal Escape. NANO LETTERS 2018; 18:6301-6311. [PMID: 30240228 DOI: 10.1021/acs.nanolett.8b02553] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Efficient small interfering RNA (siRNA) delivery in the presence of serum is of crucial importance for effective gene therapy. Fluorinated vectors are considered to be attractive candidates for siRNA-mediated gene therapy because of their delivery efficacy in serum-containing media. However, the mechanisms driving the superior gene transfection behavior of fluorinated vectors are still not well-understood, and comprehensive investigations are warranted. Herein, we fabricated a library of perfluorooctanoyl fluoride-fluorinated (PFF-fluorinated) oligoethylenimines (f xOEIs, x is the PFF:OEI feeding ratio), which can readily form nanoassemblies (f xOEI NAs) capable of efficient siRNA delivery in cells cultured in medium both devoid of and supplemented with fetal bovine serum (FBS). The gene silencing test in serum-containing medium revealed that the f0.7OEI/siRNA NAs achieved a luciferase silencing of ∼88.4% in Luc-HeLa cells cultured in FBS-containing medium, which was almost 2-fold greater than the silencing efficacy of siRNA delivered by the commercially available vector Lipo 2000 (∼48.8%). High levels of apolipoprotein B silencing were also achieved by f0.7OEI/siRNA NAs in vivo. For an assessment of the underlying mechanisms of the efficacy of gene silencing of fluorinated vectors, two alkylated OEIs, aOEI-C8 and aOEI-C12, were fabricated as controls with similar molecular structure and hydrophobicity to that of f0.7OEI, respectively. In vitro investigations showed that the superior gene delivery exhibited by f0.7OEI NAs derived from the potent endosomal disruption capability of fluorinated vectors in the presence of serum, which was essentially attributed to the serum protein adsorption resistance of the f0.7OEI NAs. Therefore, this work provides an innovative approach to siRNA delivery as well as insights into fluorine-associated serum resistance.
Collapse
Affiliation(s)
- Tingbin Zhang
- School of Chemical Engineering and Technology , Tianjin University , Tianjin 300350 , P. R. China
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science , Beijing Institute of Technology , Beijing 100081 , P. R. China
| | - Xiaowei Ma
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Ningqiang Gong
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Xiaoli Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Lu Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Xiaoxia Ye
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Bo Hu
- Advanced Research Institute of Multidisciplinary Science, School of Life Science , Beijing Institute of Technology , Beijing 100081 , P. R. China
| | - Chunhui Li
- Advanced Research Institute of Multidisciplinary Science, School of Life Science , Beijing Institute of Technology , Beijing 100081 , P. R. China
| | - Jian-Hua Tian
- School of Chemical Engineering and Technology , Tianjin University , Tianjin 300350 , P. R. China
| | - Andrea Magrini
- Department of Biomedicine and Prevention , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Jinchao Zhang
- Chemical Biology Key Laboratory of Hebei Province, College of Chemistry & Environmental Science , Hebei University , Baoding 071002 , P. R. China
| | - Weisheng Guo
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital , Guangzhou Medical University , Guangzhou 510260 , P. R. China
| | - Jin-Feng Xing
- School of Chemical Engineering and Technology , Tianjin University , Tianjin 300350 , P. R. China
| | - Massimo Bottini
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Via Montpellier 1 , 00133 Rome , Italy
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| |
Collapse
|
36
|
|
37
|
Affiliation(s)
- Ede A Rancz
- Cortical Circuits Laboratory, Francis Crick Institute, London, UK
| | - Andreas T Schaefer
- Neurophysiology of Behaviour Laboratory, Francis Crick Institute, London, UK, and the Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| |
Collapse
|