1
|
Hu PP, Zheng LL, Zhan L, Huang CZ. Imaging of Viral Genomic RNA Replication with Nanoprobes. Methods Mol Biol 2025; 2875:145-153. [PMID: 39535646 DOI: 10.1007/978-1-0716-4248-1_12] [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: 11/16/2024]
Abstract
Viruses are a great threat to human life and health. Different viruses have its unique mechanism to efficiently infect cells, and the entry process and the nucleic acid replication using cell machine are two critical processes related to the fate of virus progeny. Real-time and long-term imaging techniques can be used to thoroughly investigate the viral infection events. This chapter will present the labeling of viral genomic RNA (gRNA) replication by developing new nanoprobes, one-donor-two-acceptors one, for example, in which the synergistic coupling of multiple energy transfer strategy, so as that the journey of viruses in live cells could be monitored and imaged in real time. Methods of labeling as well as that used for fluorescent and dark field scattering imaging are outlined.
Collapse
Affiliation(s)
- Ping Ping Hu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biomedical Analytics, Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Lin Ling Zheng
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Lei Zhan
- Key Laboratory of Biomedical Analytics, Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Cheng Zhi Huang
- Key Laboratory of Biomedical Analytics, Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| |
Collapse
|
2
|
De Ávila-Arias M, Villarreal-Camacho JL, Cadena-Cruz C, Hurtado-Gómez L, Costello HM, Rodriguez A, Burgos-Florez F, Bettin A, Kararoudi MN, Muñoz A, Peeples ME, San-Juan-Vergara H. Exploring the secrets of virus entry: the first respiratory syncytial virus carrying beta lactamase. Front Microbiol 2024; 15:1339569. [PMID: 38455070 PMCID: PMC10919290 DOI: 10.3389/fmicb.2024.1339569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Background Respiratory Syncytial Virus (RSV) presents a significant health threat, especially to young children. In-depth understanding of RSV entry mechanisms is essential for effective antiviral development. This study introduces an innovative RSV variant, featuring the fusion of the beta-lactamase (BlaM) enzyme with the RSV-P phosphoprotein, providing a versatile tool for dissecting viral entry dynamics. Methods Using the AlphaFold2 algorithm, we modeled the tertiary structure of the P-BlaM chimera, revealing structural similarities with both RSV-P and BlaM. Functional assessments, utilizing flow cytometry, quantified beta-lactamase activity and GFP expression in infected bronchial epithelial cells. Western blot analysis confirmed the integrity of P-BlaM within virions. Results The modeled P-BlaM chimera exhibited structural parallels with RSV-P and BlaM. Functional assays demonstrated robust beta-lactamase activity in recombinant virions, confirming successful P-BlaM incorporation as a structural protein. Quercetin, known for its antiviral properties, impeded viral entry by affecting virion fusion. Additionally, Ulixertinib, an ERK-1/2 inhibitor, significantly curtailed viral entry, implicating ERK-1/2 pathway signaling. Conclusions Our engineered RSV-P-BlaM chimera emerges as a valuable tool, illuminating RSV entry mechanisms. Structural and functional analyses unveil potential therapeutic targets. Quercetin and Ulixertinib, identified as distinct stage inhibitors, show promise for targeted antiviral strategies. Time-of-addition assays pinpoint quercetin's specific interference stage, advancing our comprehension of RSV entry and guiding future antiviral developments.
Collapse
Affiliation(s)
- Marcio De Ávila-Arias
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Barranquilla, Colombia
| | - Jose Luis Villarreal-Camacho
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Christian Cadena-Cruz
- Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Leidy Hurtado-Gómez
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Barranquilla, Colombia
| | - Heather M. Costello
- Genomics Services Laboratory, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Alexander Rodriguez
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Barranquilla, Colombia
| | - Francisco Burgos-Florez
- Programa de regencia en farmacia, grupo de investigación creatividad e innovación tecnológica, Corporación tecnológica Indoamérica, Barranquilla, Colombia
- Escuela de Pregrado, Dirección Académica, Vicerrectoría de Sede, Universidad Nacional de Colombia, Sede La Paz, Cesar, Colombia
| | - Alfonso Bettin
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Barranquilla, Colombia
| | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Amner Muñoz
- Departamento de Química y Biología, Universidad del Norte, Barranquilla, Colombia
| | - Mark E. Peeples
- Center for Vaccines and Immunity, The Abagail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Homero San-Juan-Vergara
- Departamento de Medicina, División Ciencias de la Salud, Universidad del Norte, Barranquilla, Colombia
| |
Collapse
|
3
|
Kieser QJ, Granoski MJ, McClelland RD, Griffiths C, Bilawchuk LM, Stojic A, Elawar F, Jamieson K, Proud D, Marchant DJ. Actin cytoskeleton remodeling disrupts physical barriers to infection and presents entry receptors to respiratory syncytial virus. J Gen Virol 2023; 104:001923. [PMID: 38015055 PMCID: PMC10768689 DOI: 10.1099/jgv.0.001923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
RSV is the leading cause of infant hospitalizations and a significant cause of paediatric and geriatric morbidity worldwide. Recently, we reported that insulin-like growth factor 1 receptor (IGF1R) was a receptor for respiratory syncytial virus (RSV) in airway epithelial cells and that activation of IGF1R recruited the coreceptor, nucleolin (NCL), to the cell surface. Cilia and mucus that line the airways pose a significant barrier to viral and bacterial infection. The cortical actin cytoskeleton has been shown by others to mediate RSV entry, so we studied the roles of the RSV receptors and actin remodelling during virus entry. We found that IGF1R expression and phosphorylation were associated with the ability of RSV to infect cells. Confocal immunofluorescence imaging showed that actin projections, a hallmark of macropinocytosis, formed around viral particles 30 min after infection. Consistent with prior reports we also found that virus particles were internalized into early endosome antigen-1 positive endosomes within 90 min. Inhibiting actin polymerization significantly reduced viral titre by approximately ten-fold. Inhibiting PI3 kinase and PKCζ in stratified air-liquid interface (ALI) models of the airway epithelium had similar effects on reducing the actin remodelling observed during infection and attenuating viral entry. Actin projections were associated with NCL interacting with RSV particles resting on apical cilia of the ALIs. We conclude that macropinocytosis-like actin projections protrude through normally protective cilia and mucus layers of stratified airway epithelium that helps present the IGF1R receptor and the NCL coreceptor to RSV particles waiting at the surface.
Collapse
Affiliation(s)
- Quinten J. Kieser
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Madison J. Granoski
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Ryley D. McClelland
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Cameron Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908,, USA
| | - Leanne M. Bilawchuk
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Aleksandra Stojic
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Farah Elawar
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| | - Kyla Jamieson
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - David Proud
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - David J. Marchant
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G-2E1, Canada
| |
Collapse
|
4
|
Cadena-Cruz C, Villarreal Camacho JL, De Ávila-Arias M, Hurtado-Gomez L, Rodriguez A, San-Juan-Vergara H. Respiratory syncytial virus entry mechanism in host cells: A general overview. Mol Microbiol 2023; 120:341-350. [PMID: 37537859 DOI: 10.1111/mmi.15133] [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: 04/10/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 08/05/2023]
Abstract
Respiratory syncytial virus (RSV) is a virus that causes acute respiratory infections in neonates and older adults. To infect host cells, the attachment glycoprotein (G) interacts with a cell surface receptor. This interaction determines the specific cell types that are susceptible to infection. RSV possesses a type I fusion protein F. Type I fusion proteins are metastable when rearrangement of the prefusion F occurs; the fusion peptide is exposed transforming the protein into postfusion form. The transition between the prefusion form and its postfusion form facilitates the viral envelope and the host cell membrane to fuse, enabling the virus to enter the host cell. Understanding the entry mechanism employed by RSV is crucial for developing effective antiviral therapies. In this review, we will discuss the various types of viral fusion proteins and explore the potential entry mechanisms utilized by RSV. A deeper understanding of these mechanisms will provide valuable insights for the development of novel approaches to treat RSV infections.
Collapse
Affiliation(s)
- C Cadena-Cruz
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - J L Villarreal Camacho
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Marcio De Ávila-Arias
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Leidy Hurtado-Gomez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Alexander Rodriguez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | | |
Collapse
|
5
|
Lee JH, Chiu JHC, Ginga NJ, Ahmed T, Thouless MD, Liu Y, Takayama S. Super-resolution imaging of linearized chromatin in tunable nanochannels. NANOSCALE HORIZONS 2023; 8:1043-1053. [PMID: 37221952 DOI: 10.1039/d3nh00096f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Nanofluidic linearization and optical mapping of naked DNA have been reported in the research literature, and implemented in commercial instruments. However, the resolution with which DNA features can be resolved is still inherently limited by both Brownian motion and diffraction-limited optics. Direct analysis of native chromatin is further hampered by difficulty in electrophoretic manipulation, which is routinely used for DNA analysis. This paper describes the development of a three-layer, tunable, nanochannel system that enables non-electrophoretic linearization and immobilization of native chromatin. Furthermore, through careful selection of self-blinking fluorescent dyes and the design of the nanochannel system, we achieve direct stochastic optical reconstruction microscopy (dSTORM) super-resolution imaging of the linearized chromatin. As an initial demonstration, rDNA chromatin extracted from Tetrahymena is analyzed by multi-color imaging of total DNA, newly synthesized DNA, and newly synthesized histone H3. Our analysis reveals a relatively even distribution of newly synthesized H3 across two halves of the rDNA chromatin with palindromic symmetry, supporting dispersive nucleosome segregation. As a proof-of-concept study, our work achieves super-resolution imaging of native chromatin fibers linearized and immobilized in tunable nanochannels. It opens up a new avenue for collecting long-range and high-resolution epigenetic information as well as genetic information.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA.
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Joyce Han-Ching Chiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA.
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Nicholas J Ginga
- Department of Mechanical and Aerospace Engineering, University of Alabama in Huntsville, Huntsville, AL 35899, USA
| | - Tasdiq Ahmed
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA.
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - M D Thouless
- Department of Mechanical Engineering and Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yifan Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA.
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA.
- The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
6
|
Risso-Ballester J, Rameix-Welti MA. Spatial resolution of virus replication: RSV and cytoplasmic inclusion bodies. Adv Virus Res 2023; 116:1-43. [PMID: 37524479 DOI: 10.1016/bs.aivir.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Respiratory Syncytial Virus (RSV) is a major cause of respiratory illness in young children, elderly and immunocompromised individuals worldwide representing a severe burden for health systems. The urgent development of vaccines or specific antivirals against RSV is impaired by the lack of knowledge regarding its replication mechanisms. RSV is a negative-sense single-stranded RNA (ssRNA) virus belonging to the Mononegavirales order (MNV) which includes other viruses pathogenic to humans as Rabies (RabV), Ebola (EBOV), or measles (MeV) viruses. Transcription and replication of viral genomes occur within cytoplasmatic virus-induced spherical inclusions, commonly referred as inclusion bodies (IBs). Recently IBs were shown to exhibit properties of membrane-less organelles (MLO) arising by liquid-liquid phase separation (LLPS). Compartmentalization of viral RNA synthesis steps in viral-induced MLO is indeed a common feature of MNV. Strikingly these key compartments still remain mysterious. Most of our current knowledge on IBs relies on the use of fluorescence microscopy. The ability to fluorescently label IBs in cells has been key to uncover their dynamics and nature. The generation of recombinant viruses expressing a fluorescently-labeled viral protein and the immunolabeling or the expression of viral fusion proteins known to be recruited in IBs are some of the tools used to visualize IBs in infected cells. In this chapter, microscope techniques and the most relevant studies that have shed light on RSV IBs fundamental aspects, including biogenesis, organization and dynamics are being discussed and brought to light with the investigations carried out on other MNV.
Collapse
Affiliation(s)
| | - Marie-Anne Rameix-Welti
- Institut Pasteur, Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Paris, France; Assistance Publique des Hôpitaux de Paris, Hôpital Ambroise Paré, Laboratoire de Microbiologie, DMU15, Paris, France.
| |
Collapse
|
7
|
Xie E, Ahmad S, Smyth RP, Sieben C. Advanced fluorescence microscopy in respiratory virus cell biology. Adv Virus Res 2023; 116:123-172. [PMID: 37524480 DOI: 10.1016/bs.aivir.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Respiratory viruses are a major public health burden across all age groups around the globe, and are associated with high morbidity and mortality rates. They can be transmitted by multiple routes, including physical contact or droplets and aerosols, resulting in efficient spreading within the human population. Investigations of the cell biology of virus replication are thus of utmost importance to gain a better understanding of virus-induced pathogenicity and the development of antiviral countermeasures. Light and fluorescence microscopy techniques have revolutionized investigations of the cell biology of virus infection by allowing the study of the localization and dynamics of viral or cellular components directly in infected cells. Advanced microscopy including high- and super-resolution microscopy techniques available today can visualize biological processes at the single-virus and even single-molecule level, thus opening a unique view on virus infection. We will highlight how fluorescence microscopy has supported investigations on virus cell biology by focusing on three major respiratory viruses: respiratory syncytial virus (RSV), Influenza A virus (IAV) and SARS-CoV-2. We will review our current knowledge of virus replication and highlight how fluorescence microscopy has helped to improve our state of understanding. We will start by introducing major imaging and labeling modalities and conclude the chapter with a perspective discussion on remaining challenges and potential opportunities.
Collapse
Affiliation(s)
- Enyu Xie
- Nanoscale Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Shazeb Ahmad
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Redmond P Smyth
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany; Faculty of Medicine, University of Würzburg, Würzburg, Germany
| | - Christian Sieben
- Nanoscale Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany; Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany.
| |
Collapse
|
8
|
Cosentino G, Marougka K, Desquesnes A, Welti N, Sitterlin D, Gault E, Rameix-Welti MA. Respiratory syncytial virus ribonucleoproteins hijack microtubule Rab11 dependent transport for intracellular trafficking. PLoS Pathog 2022; 18:e1010619. [PMID: 35797399 PMCID: PMC9262236 DOI: 10.1371/journal.ppat.1010619] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/25/2022] [Indexed: 01/31/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the primary cause of severe respiratory infection in infants worldwide. Replication of RSV genomic RNA occurs in cytoplasmic inclusions generating viral ribonucleoprotein complexes (vRNPs). vRNPs then reach assembly and budding sites at the plasma membrane. However, mechanisms ensuring vRNPs transportation are unknown. We generated a recombinant RSV harboring fluorescent RNPs allowing us to visualize moving vRNPs in living infected cells and developed an automated imaging pipeline to characterize the movements of vRNPs at a high throughput. Automatic tracking of vRNPs revealed that around 10% of the RNPs exhibit fast and directed motion compatible with transport along the microtubules. Visualization of vRNPs moving along labeled microtubules and restriction of their movements by microtubule depolymerization further support microtubules involvement in vRNPs trafficking. Approximately 30% of vRNPs colocalize with Rab11a protein, a marker of the endosome recycling (ER) pathway and we observed vRNPs and Rab11-labeled vesicles moving together. Transient inhibition of Rab11a expression significantly reduces vRNPs movements demonstrating Rab11 involvement in RNPs trafficking. Finally, Rab11a is specifically immunoprecipitated with vRNPs in infected cells suggesting an interaction between Rab11 and the vRNPs. Altogether, our results strongly suggest that RSV RNPs move on microtubules by hijacking the ER pathway.
Collapse
Affiliation(s)
- Gina Cosentino
- Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Versailles, France
| | - Katherine Marougka
- Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Versailles, France
| | - Aurore Desquesnes
- Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Versailles, France
| | - Nicolas Welti
- Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Versailles, France
| | - Delphine Sitterlin
- Université Paris-Saclay, Université de Versailles St. Quentin, UMR 1173 (2I), INSERM, Versailles, France
| | - Elyanne Gault
- Université Paris-Saclay, Université de Versailles St. Quentin; UMR 1173 (2I), INSERM; Assistance Publique des Hôpitaux de Paris, Hôpital Ambroise Paré, Laboratoire de Microbiologie, DMU15; Versailles, France
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, Université de Versailles St. Quentin; UMR 1173 (2I), INSERM; Assistance Publique des Hôpitaux de Paris, Hôpital Ambroise Paré, Laboratoire de Microbiologie, DMU15; Versailles, France
- * E-mail:
| |
Collapse
|
9
|
Butola A, Coucheron DA, Szafranska K, Ahmad A, Mao H, Tinguely JC, McCourt P, Senthilkumaran P, Mehta DS, Agarwal K, Ahluwalia BS. Multimodal on-chip nanoscopy and quantitative phase imaging reveals the nanoscale morphology of liver sinusoidal endothelial cells. Proc Natl Acad Sci U S A 2021; 118:e2115323118. [PMID: 34782474 PMCID: PMC8617407 DOI: 10.1073/pnas.2115323118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/26/2022] Open
Abstract
Visualization of three-dimensional (3D) morphological changes in the subcellular structures of a biological specimen is a major challenge in life science. Here, we present an integrated chip-based optical nanoscopy combined with quantitative phase microscopy (QPM) to obtain 3D morphology of liver sinusoidal endothelial cells (LSEC). LSEC have unique morphology with small nanopores (50-300 nm in diameter) in the plasma membrane, called fenestrations. The fenestrations are grouped in discrete clusters, which are around 100 to 200 nm thick. Thus, imaging and quantification of fenestrations and sieve plate thickness require resolution and sensitivity of sub-100 nm along both the lateral and the axial directions, respectively. In chip-based nanoscopy, the optical waveguides are used both for hosting and illuminating the sample. The fluorescence signal is captured by an upright microscope, which is converted into a Linnik-type interferometer to sequentially acquire both superresolved images and phase information of the sample. The multimodal microscope provided an estimate of the fenestration diameter of 119 ± 53 nm and average thickness of the sieve plates of 136.6 ± 42.4 nm, assuming the constant refractive index of cell membrane to be 1.38. Further, LSEC were treated with cytochalasin B to demonstrate the possibility of precise detection in the cell height. The mean phase value of the fenestrated area in normal and treated cells was found to be 161 ± 50 mrad and 109 ± 49 mrad, respectively. The proposed multimodal technique offers nanoscale visualization of both the lateral size and the thickness map, which would be of broader interest in the fields of cell biology and bioimaging.
Collapse
Affiliation(s)
- Ankit Butola
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - David A Coucheron
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Karolina Szafranska
- Faculty of Health Sciences, Department of Medical Biology, Vascular Biology Research Group, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Azeem Ahmad
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Hong Mao
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Jean-Claude Tinguely
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Peter McCourt
- Faculty of Health Sciences, Department of Medical Biology, Vascular Biology Research Group, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Paramasivam Senthilkumaran
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dalip Singh Mehta
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Krishna Agarwal
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Balpreet Singh Ahluwalia
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway;
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
10
|
Cho J, Kim S, Lee H, Rah W, Cho HC, Kim NK, Bae S, Shin DH, Lee MG, Park IH, Tanaka Y, Shin E, Yi H, Han JW, Hwang PTJ, Jun HW, Park HJ, Cho K, Lee SW, Jung JK, Levit RD, Sussman MA, Harvey RP, Yoon YS. Regeneration of infarcted mouse hearts by cardiovascular tissue formed via the direct reprogramming of mouse fibroblasts. Nat Biomed Eng 2021; 5:880-896. [PMID: 34426676 PMCID: PMC8809198 DOI: 10.1038/s41551-021-00783-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts can be directly reprogrammed into cardiomyocytes, endothelial cells or smooth muscle cells. Here we report the reprogramming of mouse tail-tip fibroblasts simultaneously into cells resembling these three cell types using the microRNA mimic miR-208b-3p, ascorbic acid and bone morphogenetic protein 4, as well as the formation of tissue-like structures formed by the directly reprogrammed cells. Implantation of the formed cardiovascular tissue into the infarcted hearts of mice led to the migration of reprogrammed cells to the injured tissue, reducing regional cardiac strain and improving cardiac function. The migrated endothelial cells and smooth muscle cells contributed to vessel formation, and the migrated cardiomyocytes, which initially displayed immature characteristics, became mature over time and formed gap junctions with host cardiomyocytes. Direct reprogramming of somatic cells to make cardiac tissue may aid the development of applications in cell therapy, disease modelling and drug discovery for cardiovascular diseases.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Nam Kyun Kim
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Seongho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Dong Hoon Shin
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Yoshiaki Tanaka
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Eric Shin
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hong Yi
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, GA, USA
| | - Ji Woong Han
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick Tae Joon Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hun-Jun Park
- Division of Cardiology, Department of Internal Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyuwon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sang Wook Lee
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jae Kyung Jung
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark A Sussman
- San Diego State University Heart Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, St Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Young-Sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Blanchard EL, Braun MR, Lifland AW, Ludeke B, Noton SL, Vanover D, Zurla C, Fearns R, Santangelo PJ. Polymerase-tagged respiratory syncytial virus reveals a dynamic rearrangement of the ribonucleocapsid complex during infection. PLoS Pathog 2020; 16:e1008987. [PMID: 33031461 PMCID: PMC7575074 DOI: 10.1371/journal.ppat.1008987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/20/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022] Open
Abstract
The ribonucleocapsid complex of respiratory syncytial virus (RSV) is responsible for both viral mRNA transcription and viral replication during infection, though little is known about how this dual function is achieved. Here, we report the use of a recombinant RSV virus with a FLAG-tagged large polymerase protein, L, to characterize and localize RSV ribonucleocapsid structures during the early and late stages of viral infection. Through proximity ligation assays and super-resolution microscopy, viral RNA and proteins in the ribonucleocapsid complex were revealed to dynamically rearrange over time, particularly between 6 and 8 hours post infection, suggesting a connection between the ribonucleocapsid structure and its function. The timing of ribonucleocapsid rearrangement corresponded with an increase in RSV genome RNA accumulation, indicating that this rearrangement is likely involved with the onset of RNA replication and secondary transcription. Additionally, early overexpression of RSV M2-2 from in vitro transcribed mRNA was shown to inhibit virus infection by rearranging the ribonucleocapsid complex. Collectively, these results detail a critical understanding into the localization and activity of RSV L and the ribonucleocapsid complex during RSV infection.
Collapse
Affiliation(s)
- Emmeline L. Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Molly R. Braun
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States of America
| | - Aaron W. Lifland
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Barbara Ludeke
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States of America
| | - Sarah L. Noton
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States of America
| | - Daryll Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Rachel Fearns
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States of America
| | - Philip J. Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| |
Collapse
|
12
|
Chen L, Liang J. An overview of functional nanoparticles as novel emerging antiviral therapeutic agents. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110924. [PMID: 32409074 PMCID: PMC7195146 DOI: 10.1016/j.msec.2020.110924] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/23/2020] [Accepted: 03/31/2020] [Indexed: 01/04/2023]
Abstract
Research on highly effective antiviral drugs is essential for preventing the spread of infections and reducing losses. Recently, many functional nanoparticles have been shown to possess remarkable antiviral ability, such as quantum dots, gold and silver nanoparticles, nanoclusters, carbon dots, graphene oxide, silicon materials, polymers and dendrimers. Despite their difference in antiviral mechanism and inhibition efficacy, these functional nanoparticles-based structures have unique features as potential antiviral candidates. In this topical review, we highlight the antiviral efficacy and mechanism of these nanoparticles. Specifically, we introduce various methods for analyzing the viricidal activity of functional nanoparticles and the latest advances in antiviral functional nanoparticles. Furthermore, we systematically describe the advantages and disadvantages of these functional nanoparticles in viricidal applications. Finally, we discuss the challenges and prospects of antiviral nanostructures. This topic review covers 132 papers and will enrich our knowledge about the antiviral efficacy and mechanism of various functional nanoparticles.
Collapse
Affiliation(s)
- Lu Chen
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jiangong Liang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
13
|
Sharma KK, Marzinek JK, Tantirimudalige SN, Bond PJ, Wohland T. Single-molecule studies of flavivirus envelope dynamics: Experiment and computation. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 143:38-51. [PMID: 30223001 DOI: 10.1016/j.pbiomolbio.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022]
Abstract
Flaviviruses are simple enveloped viruses exhibiting complex structural and functional heterogeneities. Decades of research have provided crucial basic insights, antiviral medication and moderately successful gene therapy trials. The most infectious particle is, however, not always the most abundant one in a population, questioning the utility of classic ensemble-averaging virology approaches. Indeed, viral replication is often not particularly efficient, prone to errors or containing parallel routes. Here, we review different single-molecule sensitive fluorescence methods that are employed to investigate flaviviruses. In particular, we review how (i) time-resolved Förster resonance energy transfer (trFRET) was applied to probe dengue envelope conformations; (ii) FRET-fluorescence correlation spectroscopy to investigate dengue envelope intrinsic dynamics and (iii) single particle tracking to follow the path of dengue viruses in cells. We also discuss how such methods may be supported by molecular dynamics (MD) simulations over a range of spatio-temporal scales, to provide complementary data on the structure and dynamics of flaviviral systems. We describe recent improvements in multiscale MD approaches that allowed the simulation of dengue particle envelopes in near-atomic resolution. We hope this review is an incentive for setting up and applying similar single-molecule studies and combine them with MD simulations to investigate structural dynamics of entire flavivirus particles over the nanosecond-to-millisecond time-scale and follow viruses during infection in cells over milliseconds to minutes.
Collapse
Affiliation(s)
- Kamal Kant Sharma
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Jan K Marzinek
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Sarala Neomi Tantirimudalige
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore
| | - Peter J Bond
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore.
| | - Thorsten Wohland
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Department of Chemistry, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; Centre for Bioimaging Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117557, Singapore.
| |
Collapse
|
14
|
Loomis KH, Lindsay KE, Zurla C, Bhosle SM, Vanover DA, Blanchard EL, Kirschman JL, Bellamkonda RV, Santangelo PJ. In Vitro Transcribed mRNA Vaccines with Programmable Stimulation of Innate Immunity. Bioconjug Chem 2018; 29:3072-3083. [PMID: 30067354 DOI: 10.1021/acs.bioconjchem.8b00443] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In vitro transcribed (IVT) mRNA is an appealing platform for next generation vaccines, as it can be manufactured rapidly at large scale to meet emerging pathogens. However, its performance as a robust vaccine is strengthened by supplemental immune stimulation, which is typically provided by adjuvant formulations that facilitate delivery and stimulate immune responses. Here, we present a strategy for increasing translation of a model IVT mRNA vaccine while simultaneously modulating its immune-stimulatory properties in a programmable fashion, without relying on delivery vehicle formulations. Substitution of uridine with the modified base N1-methylpseudouridine reduces the intrinsic immune stimulation of the IVT mRNA and enhances antigen translation. Tethering adjuvants to naked IVT mRNA through antisense nucleotides boosts the immunostimulatory properties of adjuvants in vitro, without impairing transgene production or adjuvant activity. In vivo, intramuscular injection of tethered IVT mRNA-TLR7 agonists leads to enhanced local immune responses, and to antigen-specific cell-mediated and humoral responses. We believe this system represents a potential platform compatible with any adjuvant of interest to enable specific programmable stimulation of immune responses.
Collapse
Affiliation(s)
- Kristin H Loomis
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Kevin E Lindsay
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Sushma M Bhosle
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Emmeline L Blanchard
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Jonathan L Kirschman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Ravi V Bellamkonda
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University , Krone Engineering Biosystems Building, 950 Atlantic Drive , Atlanta , Georgia 30332 , United States
| |
Collapse
|
15
|
Unifying in vitro and in vivo IVT mRNA expression discrepancies in skeletal muscle via mechanotransduction. Biomaterials 2018; 159:189-203. [PMID: 29331806 DOI: 10.1016/j.biomaterials.2018.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/30/2017] [Accepted: 01/06/2018] [Indexed: 12/23/2022]
Abstract
The translational efficiency of an in vitro transcribed (IVT) mRNA was measured upon delivery to primary skeletal muscle cells and to a mouse model system, towards the development of a predictive in vitro assay for the screening and validation of intramuscular mRNA-based vaccines. When IVT mRNA was delivered either naked or complexed with novel aminoglycoside-based delivery vehicles, significant differences in protein expression in vitro and in vivo were observed. We hypothesized that this previously anticipated discrepancy was due to differences in the mechanism of IVT mRNA endosomal entry and release following delivery. To address this, IVT mRNA was fluorescently labeled prior to delivery, to visualize its distribution. Colocalization with endosomal markers indicated that different entry pathways were utilized in vivo and in vitro, depending on the delivery vehicle, resulting in variations in protein expression levels. Since extracellular matrix stiffness (ECM) influences mRNA entry, trafficking and release, the effect of mechanotransduction on mRNA expression was investigated in vitro upon delivery of IVT mRNA alone, and complexed with delivery vehicles to skeletal muscle cells grown on ∼10 kPa hydrogels. This in vitro hydrogel model more accurately recapitulated the results obtained in vivo upon IM injection, indicating that this approach may assist in the characterization of mRNA based vaccines.
Collapse
|
16
|
Abstract
The last past decade has witnessed a revolution in our appreciation of transcriptome complexity and regulation. This remarkable expansion in our knowledge largely originates from the advent of high-throughput methodologies, and the consecutive discovery that up to 90% of eukaryotic genomes are transcribed, thus generating an unanticipated large range of noncoding RNAs (Hangauer et al., 15(4):112, 2014). Besides leading to the identification of new noncoding RNA species, transcriptome-wide studies have uncovered novel layers of posttranscriptional regulatory mechanisms controlling RNA processing, maturation or translation, and each contributing to the precise and dynamic regulation of gene expression. Remarkably, the development of systems-level studies has been accompanied by tremendous progress in the visualization of individual RNA molecules in single cells, such that it is now possible to image RNA species with a single-molecule resolution from birth to translation or decay. Monitoring quantitatively, with unprecedented spatiotemporal resolution, the fate of individual molecules has been key to understanding the molecular mechanisms underlying the different steps of RNA regulation. This has also revealed biologically relevant, intracellular and intercellular heterogeneities in RNA distribution or regulation. More recently, the convergence of imaging and high-throughput technologies has led to the emergence of spatially resolved transcriptomic techniques that provide a means to perform large-scale analyses while preserving spatial information. By generating transcriptome-wide data on single-cell RNA content, or even subcellular RNA distribution, these methodologies are opening avenues to a wide range of network-level studies at the cell and organ-level, and promise to strongly improve disease diagnostic and treatment.In this introductory chapter, we highlight how recently developed technologies aiming at detecting and visualizing RNA molecules have contributed to the emergence of entirely new research fields, and to dramatic progress in our understanding of gene expression regulation.
Collapse
Affiliation(s)
- Caroline Medioni
- Université Côte d'Azur, CNRS, Inserm, iBV, Parc Valrose, 06100, Nice, France
| | - Florence Besse
- Université Côte d'Azur, CNRS, Inserm, iBV, Parc Valrose, 06100, Nice, France.
| |
Collapse
|
17
|
RSV glycoprotein and genomic RNA dynamics reveal filament assembly prior to the plasma membrane. Nat Commun 2017; 8:667. [PMID: 28939853 PMCID: PMC5610308 DOI: 10.1038/s41467-017-00732-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 07/20/2017] [Indexed: 11/25/2022] Open
Abstract
The human respiratory syncytial virus G protein plays an important role in the entry and assembly of filamentous virions. Here, we report the use of fluorescently labeled soybean agglutinin to selectively label the respiratory syncytial virus G protein in living cells without disrupting respiratory syncytial virus infectivity or filament formation and allowing for interrogations of respiratory syncytial virus virion assembly. Using this approach, we discovered that plasma membrane-bound respiratory syncytial virus G rapidly recycles from the membrane via clathrin-mediated endocytosis. This event is then followed by the dynamic formation of filamentous and branched respiratory syncytial virus particles, and assembly with genomic ribonucleoproteins and caveolae-associated vesicles prior to re-insertion into the plasma membrane. We demonstrate that these processes are halted by the disruption of microtubules and inhibition of molecular motors. Collectively, our results show that for respiratory syncytial virus assembly, viral filaments are produced and loaded with genomic RNA prior to insertion into the plasma membrane. Assembly of filamentous RSV particles is incompletely understood due to a lack of techniques suitable for live-cell imaging. Here Vanover et al. use labeled soybean agglutinin to selectively label RSV G protein and show how filamentous RSV assembly, initiated in the cytoplasm, uses G protein recycled from the plasma membrane.
Collapse
|
18
|
Kirschman JL, Bhosle S, Vanover D, Blanchard EL, Loomis KH, Zurla C, Murray K, Lam BC, Santangelo PJ. Characterizing exogenous mRNA delivery, trafficking, cytoplasmic release and RNA-protein correlations at the level of single cells. Nucleic Acids Res 2017; 45:e113. [PMID: 28449134 PMCID: PMC5499550 DOI: 10.1093/nar/gkx290] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/06/2017] [Accepted: 04/11/2017] [Indexed: 12/25/2022] Open
Abstract
The use of synthetic messenger ribonucleic acid (mRNA) to express specific proteins is a highly promising therapeutic and vaccine approach that avoids many safety issues associated with viral or DNA-based systems. However, in order to optimize mRNA designs and delivery, technology advancements are required to study fundamental mechanisms of mRNA uptake and localization at the single-cell and tissue level. Here, we present a single RNA sensitive fluorescent labeling method which allows us to label and visualize synthetic mRNA without significantly affecting function. This approach enabled single cell characterization of mRNA uptake and release kinetics from endocytic compartments, the measurement of mRNA/protein correlations, and motivated the investigation of mRNA induced cellular stress, all important mechanisms influencing protein production. In addition, we demonstrated this approach can facilitate near-infrared imaging of mRNA localization in vivo and in ex-vivo tissue sections, which will facilitate mRNA trafficking studies in pre-clinical models. Overall, we demonstrate the ability to study fundamental mechanisms necessary to optimize delivery and therapeutic strategies, in order to design the next generation of novel mRNA therapeutics and vaccines.
Collapse
Affiliation(s)
- Jonathan L. Kirschman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Sushma Bhosle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Daryll Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Emmeline L. Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Kristin H. Loomis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Kathryn Murray
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Blaine C. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Philip J. Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, UA Whitaker Building, 313 Ferst Drive, Atlanta, GA 30332, USA
| |
Collapse
|
19
|
Techniques for Single-Molecule mRNA Imaging in Living Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:425-441. [DOI: 10.1007/978-3-319-53889-1_22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
A Novel Method to Quantify RNA-Protein Interactions In Situ Using FMTRIP and Proximity Ligation. Methods Mol Biol 2017; 1468:155-70. [PMID: 27662876 DOI: 10.1007/978-1-4939-4035-6_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RNA binding proteins (RBP) and small RNAs regulate the editing, localization, stabilization, translation, and degradation of ribonucleic acids (RNAs) through their interactions with specific cis-acting elements within target RNAs. Here, we describe a novel method to detect protein-mRNA interactions, which combines FLAG-peptide modified, multiply-labeled tetravalent RNA imaging probes (FMTRIPs) with proximity ligation (PLA), and rolling circle amplification (RCA). This assay detects native RNA in a sequence specific and single RNA sensitive manner, and PLA allows for the quantification and localization of protein-mRNA interactions with single-interaction sensitivity.
Collapse
|
21
|
Ferrer M, Henriet S, Chamontin C, Lainé S, Mougel M. From Cells to Virus Particles: Quantitative Methods to Monitor RNA Packaging. Viruses 2016; 8:v8080239. [PMID: 27556480 PMCID: PMC4997601 DOI: 10.3390/v8080239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
In cells, positive strand RNA viruses, such as Retroviridae, must selectively recognize their full-length RNA genome among abundant cellular RNAs to assemble and release particles. How viruses coordinate the intracellular trafficking of both RNA and protein components to the assembly sites of infectious particles at the cell surface remains a long-standing question. The mechanisms ensuring packaging of genomic RNA are essential for viral infectivity. Since RNA packaging impacts on several essential functions of retroviral replication such as RNA dimerization, translation and recombination events, there are many studies that require the determination of RNA packaging efficiency and/or RNA packaging ability. Studies of RNA encapsidation rely upon techniques for the identification and quantification of RNA species packaged by the virus. This review focuses on the different approaches available to monitor RNA packaging: Northern blot analysis, ribonuclease protection assay and quantitative reverse transcriptase-coupled polymerase chain reaction as well as the most recent RNA imaging and sequencing technologies. Advantages, disadvantages and limitations of these approaches will be discussed in order to help the investigator to choose the most appropriate technique. Although the review was written with the prototypic simple murine leukemia virus (MLV) and complex human immunodeficiency virus type 1 (HIV-1) in mind, the techniques were described in order to benefit to a larger community.
Collapse
Affiliation(s)
- Mireia Ferrer
- CPBS, CNRS, Université de Montpellier, 1919 Route de Mende, Montpellier 34293, France.
| | - Simon Henriet
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen 5018, Norway.
| | - Célia Chamontin
- CPBS, CNRS, Université de Montpellier, 1919 Route de Mende, Montpellier 34293, France.
| | - Sébastien Lainé
- CPBS, CNRS, Université de Montpellier, 1919 Route de Mende, Montpellier 34293, France.
| | - Marylène Mougel
- CPBS, CNRS, Université de Montpellier, 1919 Route de Mende, Montpellier 34293, France.
| |
Collapse
|
22
|
Alonas E, Vanover D, Blanchard E, Zurla C, Santangelo PJ. Imaging viral RNA using multiply labeled tetravalent RNA imaging probes in live cells. Methods 2016; 98:91-98. [PMID: 26875782 DOI: 10.1016/j.ymeth.2016.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/05/2023] Open
Abstract
Viruses represent an important class of pathogens that have had an enormous impact on the health of the human race. They are extraordinarily diverse; viral particles can range in size from ∼80nm to ∼10μm in length, and contain genomes with RNA or DNA strands. Regardless of their genome type, RNA species are frequently generated as a part of their replication process, and for viruses with RNA genomes, their loading into the virion represents a critical step in the creation of infectious particles. RNA imaging tools represent a powerful approach to gain insight into fundamental viral processes, including virus entry, replication, and virion assembly. Imaging viral processes in live cells is critical due to both the heterogeneity of these processes on a per cell basis, and the inherent dynamics of these processes. There are a number of methods for labeling RNA in live cells; we'll introduce the myriad of methods and then focus on one approach for labeling viral RNA, using multiply-labeled tetravalent RNA imaging probes (MTRIPs), which do not require engineering of the target RNAs. We feel this approach is advantageous given many viral genomes may not tolerate large nucleotide insertions into their sequences.
Collapse
Affiliation(s)
- Eric Alonas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Daryll Vanover
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Emmeline Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, United States.
| |
Collapse
|
23
|
Zurla C, Jung J, Santangelo PJ. Can we observe changes in mRNA "state"? Overview of methods to study mRNA interactions with regulatory proteins relevant in cancer related processes. Analyst 2016; 141:548-62. [PMID: 26605378 PMCID: PMC4701657 DOI: 10.1039/c5an01959a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RNA binding proteins (RBP) regulate the editing, localization, stabilization, translation, and degradation of ribonucleic acids (RNA) through their interactions with specific cis-acting elements within target RNAs. Post-transcriptional regulatory mechanisms are directly involved in the control of the immune response and stress response and their alterations play a crucial role in cancer related processes. In this review, we discuss mRNAs and RNA binding proteins relevant to tumorigenesis, current methodologies for detecting RNA interactions, and last, we describe a novel method to detect such interactions, which combines peptide modified, RNA imaging probes (FMTRIPs) with proximity ligation (PLA) and rolling circle amplification (RCA). This assay detects native RNA in a sequence specific and single RNA sensitive manner, and PLA allows for the quantification and localization of protein-mRNA interactions with single-interaction sensitivity in situ.
Collapse
Affiliation(s)
- C Zurla
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Blgd, Atlanta, GA 30332, USA.
| | - J Jung
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Blgd, Atlanta, GA 30332, USA.
| | - P J Santangelo
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, UA Whitaker Blgd, Atlanta, GA 30332, USA.
| |
Collapse
|
24
|
Yang X, Xie H, Alonas E, Liu Y, Chen X, Santangelo PJ, Ren Q, Xi P, Jin D. Mirror-enhanced super-resolution microscopy. LIGHT, SCIENCE & APPLICATIONS 2016; 5. [PMID: 27398242 PMCID: PMC4936537 DOI: 10.1038/lsa.2016.134] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Axial excitation confinement beyond the diffraction limit is crucial to the development of next-generation, super-resolution microscopy. STimulated Emission Depletion (STED) nanoscopy offers lateral super-resolution using a donut-beam depletion, but its axial resolution is still over 500 nm. Total internal reflection fluorescence microscopy is widely used for single-molecule localization, but its ability to detect molecules is limited to within the evanescent field of ~ 100 nm from the cell attachment surface. We find here that the axial thickness of the point spread function (PSF) during confocal excitation can be easily improved to 110 nm by replacing the microscopy slide with a mirror. The interference of the local electromagnetic field confined the confocal PSF to a 110-nm spot axially, which enables axial super-resolution with all laser-scanning microscopes. Axial sectioning can be obtained with wavelength modulation or by controlling the spacer between the mirror and the specimen. With no additional complexity, the mirror-assisted excitation confinement enhanced the axial resolution six-fold and the lateral resolution two-fold for STED, which together achieved 19-nm resolution to resolve the inner rim of a nuclear pore complex and to discriminate the contents of 120 nm viral filaments. The ability to increase the lateral resolution and decrease the thickness of an axial section using mirror-enhanced STED without increasing the laser power is of great importance for imaging biological specimens, which cannot tolerate high laser power.
Collapse
Affiliation(s)
- Xusan Yang
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
| | - Hao Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Eric Alonas
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Yujia Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
- Advanced Cytometry Labs, ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney, NSW 2109, Australia
| | - Xuanze Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
| | - Philip J Santangelo
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
| | - Peng Xi
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Beijing 100871, China
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Advanced Cytometry Labs, ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney, NSW 2109, Australia
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Dayong Jin
- Advanced Cytometry Labs, ARC Centre of Excellence for Nanoscale BioPhotonics (CNBP), Macquarie University, Sydney, NSW 2109, Australia
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
25
|
Yi H, Strauss JD, Ke Z, Alonas E, Dillard RS, Hampton CM, Lamb KM, Hammonds JE, Santangelo PJ, Spearman PW, Wright ER. Native immunogold labeling of cell surface proteins and viral glycoproteins for cryo-electron microscopy and cryo-electron tomography applications. J Histochem Cytochem 2015; 63:780-92. [PMID: 26069287 PMCID: PMC4823802 DOI: 10.1369/0022155415593323] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/29/2015] [Indexed: 11/22/2022] Open
Abstract
Numerous methods have been developed for immunogold labeling of thick, cryo-preserved biological specimens. However, most of the methods are permutations of chemical fixation and sample sectioning, which select and isolate the immunolabeled region of interest. We describe a method for combining immunogold labeling with cryo-electron microscopy (cryo-EM) and cryo-electron tomography (cryo-ET) of the surface proteins of intact mammalian cells or the surface glycoproteins of assembling and budding viruses in the context of virus-infected mammalian cells cultured on EM grids. In this method, the cells were maintained in culture media at physiologically relevant temperatures while sequentially incubated with the primary and secondary antibodies. Subsequently, the immunogold-labeled specimens were vitrified and observed under cryo-conditions in the transmission electron microscope. Cryo-EM and cryo-ET examination of the immunogold-labeled cells revealed the association of immunogold particles with the target antigens. Additionally, the cellular structure was unaltered by pre-immunolabeling chemical fixation and retained well-preserved plasma membranes, cytoskeletal elements, and macromolecular complexes. We think this technique will be of interest to cell biologists for cryo-EM and conventional studies of native cells and pathogen-infected cells.
Collapse
Affiliation(s)
- Hong Yi
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, Georgia (HY, ERW)
| | - Joshua D Strauss
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Zunlong Ke
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia (ZK)
| | - Eric Alonas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia (EA, PJS)
| | - Rebecca S Dillard
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Cheri M Hampton
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Kristen M Lamb
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Jason E Hammonds
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia (EA, PJS)
| | - Paul W Spearman
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| | - Elizabeth R Wright
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, Georgia (HY, ERW)
- Division of Pediatric Infectious Diseases, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia (JDS, RSD, CMH, KML, JEH, PWS, ERW)
| |
Collapse
|
26
|
Abstract
The localization of mRNAs to specific subcellular sites is widespread, allowing cells to spatially restrict and regulate protein production, and playing important roles in development and cellular physiology. This process has been studied in mechanistic detail for several RNAs. However, the generality or specificity of RNA localization systems and mechanisms that impact the many thousands of localized mRNAs has been difficult to assess. In this review, we discuss the current state of the field in determining which RNAs localize, which RNA sequences mediate localization, the protein factors involved, and the biological implications of localization. For each question, we examine prominent systems and techniques that are used to study individual messages, highlight recent genome-wide studies of RNA localization, and discuss the potential for adapting other high-throughput approaches to the study of localization.
Collapse
Affiliation(s)
- J Matthew Taliaferro
- a Department of Biology; Massachusetts Institute of Technology ; Cambridge , MA USA
| | | | | |
Collapse
|
27
|
Zheng LL, Yang XX, Liu Y, Wan XY, Wu WB, Wang TT, Wang Q, Zhen SJ, Huang CZ. In situ labelling chemistry of respiratory syncytial viruses by employing the biotinylated host-cell membrane protein for tracking the early stage of virus entry. Chem Commun (Camb) 2014; 50:15776-9. [PMID: 25370508 DOI: 10.1039/c4cc06264g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An in situ labelling strategy was proposed to produce quantum dot-labelled respiratory syncytial viruses (RSVs) by incorporating the biotinylated membrane protein of the host cells into mature virions, followed by conjugation with streptavidin modified quantum dots (SA-QDs), which has the advantages such as convenience, efficiency and minor influence on viral infectivity and thus could be successfully applied to track the early stage of virus entry.
Collapse
Affiliation(s)
- Lin Ling Zheng
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jensen E, Crossman DJ. Technical review: types of imaging-direct STORM. Anat Rec (Hoboken) 2014; 297:2227-31. [PMID: 24995970 DOI: 10.1002/ar.22960] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/09/2014] [Indexed: 12/22/2022]
Abstract
In the 1990s, new concepts of microscopy revolutionized the imaging field by breaking the lateral resolution diffraction limit for the first time, even with propagating light and regular lenses (i.e., far-field). In 2006, several research groups independently showed super-resolution microscopy using high-precision localization of single fluorophores. These new developments in single-molecule spectroscopy enabled a different approach to achieving nanometer-scale optical microscopy. Direct stochastic optical reconstruction microscopy (dSTORM) is a technique of single-molecule super-resolution imaging that does not require an activator fluorophore. This technique is used to visualize cellular structures with a resolution of approximately 20 nm. dSTORM is compatible with many conventionally used fluorophores. This article provides an overview of the principles and uses of dSTORM. Advantages and disadvantages of dSTORM are also discussed.
Collapse
|