Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38260360

ABSTRACT

Understanding the spatial organization of nucleoporins (Nups) with intrinsically disordered domains within the nuclear pore complex (NPC) is crucial for deciphering eukaryotic nucleocytoplasmic transport. Leveraging high-speed 2D single-molecule tracking and virtual 3D super-resolution microscopy in live HeLa cells, we investigated the spatial distribution of all eleven phenylalanine-glycine (FG)-rich Nups within individual NPCs. Our study reveals a nuanced landscape of FG-Nup conformations and arrangements. Five FG-Nups are steadfastly anchored at the NPC scaffold, collectively shaping a central doughnut-shaped channel, while six others exhibit heightened flexibility, extending towards the cytoplasmic and nucleoplasmic regions. Intriguingly, Nup214 and Nup153 contribute to cap-like structures that dynamically alternate between open and closed states along the nucleocytoplasmic transport axis, impacting the cytoplasmic and nuclear sides, respectively. Furthermore, Nup98, concentrated at the scaffold region, extends throughout the entire NPC while overlapping with other FG-Nups. Together, these eleven FG-Nups compose a versatile, capped trichoid channel spanning approximately 270 nm across the nuclear envelope. This adaptable trichoid channel facilitates a spectrum of pathways for passive diffusion and facilitated nucleocytoplasmic transport. Our comprehensive mapping of FG-Nup organization within live NPCs offers a unifying mechanism accommodating multiple transport pathways, thereby advancing our understanding of cellular transport processes.

2.
STAR Protoc ; 5(1): 102790, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38113144

ABSTRACT

Here, we present a protocol for single-molecule super-resolution imaging of the nuclear export of pre-ribosomal subunits pre-40S and pre-60S through nuclear pore complexes. We describe steps for plating cells and co-transfecting cells. We then detail steps for using single-point edge-excitation sub-diffraction microscopy, allowing visualization of real-time dynamics of the pre-ribosomal subunits. For complete details on the use and execution of this protocol, please refer to Junod et al. (2023).1.


Subject(s)
Nuclear Pore , Saccharomyces cerevisiae Proteins , Nuclear Pore/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Active Transport, Cell Nucleus , Ribosome Subunits/metabolism , Single Molecule Imaging/methods
3.
iScience ; 26(8): 107445, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37599825

ABSTRACT

We present a study on the nuclear export efficiency and time of pre-ribosomal subunits in live mammalian cells, using high-speed single-molecule tracking and single-molecule fluorescence resonance energy transfer techniques. Our findings reveal that pre-ribosomal particles exhibit significantly higher nuclear export efficiency compared to other large cargos like mRNAs, with around two-thirds of interactions between the pre-60S or pre-40S and the nuclear pore complexes (NPCs) resulting in successful export to the cytoplasm. We also demonstrate that nuclear transport receptor (NTR) chromosomal maintenance 1 (CRM1) plays a crucial role in nuclear export efficiency, with pre-60S and pre-40S particle export efficiency decreasing by 11-17-fold when CRM1 is inhibited. Our results suggest that multiple copies of CRM1 work cooperatively to chaperone pre-ribosomal subunits through the NPC, thus increasing export efficiency and decreasing export time. Significantly, this cooperative NTR mechanism extends beyond pre-ribosomal subunits, as evidenced by the enhanced nucleocytoplasmic transport of proteins.

4.
Chem Biomed Imaging ; 1(4): 356-371, 2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37501792

ABSTRACT

Super-resolution imaging techniques have broken the diffraction-limited resolution of light microscopy. However, acquiring three-dimensional (3D) super-resolution information about structures and dynamic processes in live cells at high speed remains challenging. Recently, the development of high-speed single-point edge-excitation subdiffraction (SPEED) microscopy, along with its 2D-to-3D transformation algorithm, provides a practical and effective approach to achieving 3D subdiffraction-limit information in subcellular structures and organelles with rotational symmetry. One of the major benefits of SPEED microscopy is that it does not rely on complex optical components and can be implemented on a standard, inverted epifluorescence microscope, simplifying the process of sample preparation and the expertise requirement. SPEED microscopy is specifically designed to obtain 2D spatial locations of individual immobile or moving fluorescent molecules inside submicrometer biological channels or cavities at high spatiotemporal resolution. The collected data are then subjected to postlocalization 2D-to-3D transformation to obtain 3D super-resolution structural and dynamic information. In recent years, SPEED microscopy has provided significant insights into nucleocytoplasmic transport across the nuclear pore complex (NPC) and cytoplasm-cilium trafficking through the ciliary transition zone. This Review focuses on the applications of SPEED microscopy in studying the structure and function of nuclear pores.

5.
Comput Struct Biotechnol J ; 21: 1424-1432, 2023.
Article in English | MEDLINE | ID: mdl-36824228

ABSTRACT

Super-resolution imaging techniques have provided unprecedentedly detailed information by surpassing the diffraction-limited resolution of light microscopy. However, in order to derive high quality spatial resolution, many of these techniques require high laser power, extended imaging time, dedicated sample preparation, or some combination of the three. These constraints are particularly evident when considering three-dimensional (3D) super-resolution imaging. As a result, high-speed capture of 3D super-resolution information of structures and dynamic processes within live cells remains both desirable and challenging. Recently, a highly effective approach to obtain 3D super-resolution information was developed that can be employed in commonly available laboratory microscopes. This development makes it both scientifically possible and financially feasible to obtain super-resolution 3D information under certain conditions. This is accomplished by converting 2D single-molecule localization data captured at high speed within subcellular structures and rotationally symmetric organelles. Here, a high-speed 2D single-molecule tracking and post-localization technique, known as single-point edge-excitation sub-diffraction (SPEED) microcopy, along with its 2D-to-3D transformation algorithm is detailed with special emphasis on the mathematical principles and Monte Carlo simulation validation of the technique.

6.
Cells ; 11(19)2022 09 30.
Article in English | MEDLINE | ID: mdl-36231040

ABSTRACT

The transient nature of RNA has rendered it one of the more difficult biological targets for imaging. This difficulty stems both from the physical properties of RNA as well as the temporal constraints associated therewith. These concerns are further complicated by the difficulty in imaging endogenous RNA within a cell that has been transfected with a target sequence. These concerns, combined with traditional concerns associated with super-resolution light microscopy has made the imaging of this critical target difficult. Recent advances have provided researchers the tools to image endogenous RNA in live cells at both the cellular and single-molecule level. Here, we review techniques used for labeling and imaging RNA with special emphases on various labeling methods and a virtual 3D super-resolution imaging technique.


Subject(s)
Imaging, Three-Dimensional , Single Molecule Imaging , Imaging, Three-Dimensional/methods , RNA , RNA, Messenger/genetics , Single Molecule Imaging/methods
7.
Nat Commun ; 13(1): 350, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039490

ABSTRACT

We report the discovery of a facile peptide macrocyclization and stapling strategy based on a fluorine thiol displacement reaction (FTDR), which renders a class of peptide analogues with enhanced stability, affinity, cellular uptake, and inhibition of cancer cells. This approach enabled selective modification of the orthogonal fluoroacetamide side chains in unprotected peptides in the presence of intrinsic cysteines. The identified benzenedimethanethiol linker greatly promoted the alpha helicity of a variety of peptide substrates, as corroborated by molecular dynamics simulations. The cellular uptake of benzenedimethanethiol stapled peptides appeared to be universally enhanced compared to the classic ring-closing metathesis (RCM) stapled peptides. Pilot mechanism studies suggested that the uptake of FTDR-stapled peptides may involve multiple endocytosis pathways in a distinct pattern in comparison to peptides stapled by RCM. Consistent with the improved cell permeability, the FTDR-stapled lead Axin and p53 peptide analogues demonstrated enhanced inhibition of cancer cells over the RCM-stapled analogues and the unstapled peptides.


Subject(s)
Fluorine/chemistry , Macrocyclic Compounds/chemistry , Peptides/chemistry , Sulfhydryl Compounds/chemistry , Amino Acid Sequence , Axin Protein/chemistry , Cell Membrane Permeability , Cell-Penetrating Peptides/chemistry , Cross-Linking Reagents/chemistry , Cyclization , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Dynamics Simulation , Thermodynamics , Tumor Suppressor Protein p53/chemistry
8.
Viruses ; 13(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499411

ABSTRACT

Understanding the detailed nuclear import kinetics of adeno-associated virus (AAV) through the nuclear pore complex (NPC) is essential for the application of AAV capsids as a nuclear delivery instrument as well as a target for drug development. However, a comprehensive understanding of AAV transport through the sub-micrometer NPCs in live cells calls for new techniques that can conquer the limitations of conventional fluorescence microscopy and electron microscopy. With recent technical advances in single-molecule fluorescence microscopy, we are now able to image the entire nuclear import process of AAV particles and also quantify the transport dynamics of viral particles through the NPCs in live human cells. In this review, we initially evaluate the necessity of single-molecule live-cell microscopy in the study of nuclear import for AAV particles. Then, we detail the application of high-speed single-point edge-excitation sub-diffraction (SPEED) microscopy in tracking the entire process of nuclear import for AAV particles. Finally, we summarize the major findings for AAV nuclear import by using SPEED microscopy.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus/virology , Dependovirus/metabolism , Nuclear Pore/virology , Single Molecule Imaging , Capsid/metabolism , Cell Line , Cell Nucleus/metabolism , Humans , Microscopy, Fluorescence , Nuclear Pore/metabolism
9.
Nat Commun ; 11(1): 2184, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366843

ABSTRACT

Roughly 10% of eukaryotic transmembrane proteins are found on the nuclear membrane, yet how such proteins target and translocate to the nucleus remains in dispute. Most models propose transport through the nuclear pore complexes, but a central outstanding question is whether transit occurs through their central or peripheral channels. Using live-cell high-speed super-resolution single-molecule microscopy we could distinguish protein translocation through the central and peripheral channels, finding that most inner nuclear membrane proteins use only the peripheral channels, but some apparently extend intrinsically disordered domains containing nuclear localization signals into the central channel for directed nuclear transport. These nucleoplasmic signals are critical for central channel transport as their mutation blocks use of the central channels; however, the mutated proteins can still complete their translocation using only the peripheral channels, albeit at a reduced rate. Such proteins can still translocate using only the peripheral channels when central channel is blocked, but blocking the peripheral channels blocks translocation through both channels. This suggests that peripheral channel transport is the default mechanism that was adapted in evolution to include aspects of receptor-mediated central channel transport for directed trafficking of certain membrane proteins.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Active Transport, Cell Nucleus , Fluorescence Recovery After Photobleaching , HeLa Cells , Humans , Luminescent Proteins/metabolism , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Nucleocytoplasmic Transport Proteins/metabolism , Protein Transport
10.
Protein Sci ; 29(6): 1459-1472, 2020 06.
Article in English | MEDLINE | ID: mdl-32096308

ABSTRACT

Both natively folded and intrinsically disordered proteins (IDPs) destined for the nucleus need to transport through the nuclear pore complexes (NPCs) in eukaryotic cells. NPCs allow for passive diffusion of small folded proteins while barricading large ones, unless they are facilitated by nuclear transport receptors. However, whether nucleocytoplasmic transport of IDPs would follow these rules remains unknown. By using a high-speed super-resolution fluorescence microscopy, we have measured transport kinetics and 3D spatial locations of transport routes through native NPCs for various IDPs. Our data revealed that the rules executed for folded proteins are not well followed by the IDPs. Instead, both large and small IDPs can passively diffuse through the NPCs. Furthermore, their diffusion efficiencies and routes are differentiated by their content ratio of charged (Ch) and hydrophobic (Hy) amino acids. A Ch/Hy-ratio mechanism was finally suggested for nucleocytoplasmic transport of IDPs.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Intrinsically Disordered Proteins/metabolism , Active Transport, Cell Nucleus , Eukaryotic Cells/metabolism , HeLa Cells , Humans , Kinetics , Microscopy, Fluorescence , Nuclear Pore/metabolism , Tumor Cells, Cultured
11.
Methods ; 153: 46-62, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30125665

ABSTRACT

The nuclear exit of messenger RNA (mRNA) molecules through the nuclear pore complex (NPC) is an essential step in the translation process of all proteins. The current limitations of conventional fluorescence and electron microscopy have prevented elucidation of how mRNA exports through the NPCs of live cells. In the recent years, various single-molecule fluorescence (SMF) microscopy techniques have been developed to improve the temporal and spatial resolutions of live-cell imaging allowing a more comprehensive understanding of the dynamics of mRNA export through native NPCs. In this review, we firstly evaluate the necessity of single-molecule live-cell microscopy in the study of mRNA nuclear export. Then, we highlight the application of single-point edge-excitation sub-diffraction (SPEED) microscopy that combines high-speed SMF microscopy and a 2D-to-3D transformation algorithm in the studies of nuclear transport kinetics and route for mRNAs. Finally, we summarize the new features of mRNA nuclear export found with SPEED microscopy as well as the reliability and accuracy of SPEED microscopy in mapping the 3D spatial locations of transport routes adopted by proteins and mRNAs through the NPCs.


Subject(s)
Active Transport, Cell Nucleus , Nuclear Pore/metabolism , RNA, Messenger/metabolism , Single Molecule Imaging/methods , Algorithms , Animals , Eukaryota/metabolism , Humans , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Kinetics , Microscopy, Fluorescence/methods , Ribonucleoproteins/metabolism
12.
J Cell Sci ; 130(7): 1299-1306, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28202688

ABSTRACT

The nuclear pore complex (NPC), composed of ∼30 different nucleoporins (Nups), is one of the largest supramolecular structures in eukaryotic cells. Its octagonal ring scaffold perforates the nuclear envelope and features a unique molecular machinery that regulates nucleocytoplasmic transport. However, the precise copy number and the spatial location of each Nup in the native NPC remain obscure due to the inherent difficulty of counting and localizing proteins inside of the sub-micrometer supramolecular complex. Here, we combined super-resolution single-point edge-excitation subdiffraction (SPEED) microscopy and nanobody-specific labeling to reveal the spatial distribution of scaffold Nups within three separate layers in the native NPC with a precision of ∼3 nm. Our data reveal both the radial and axial spatial distributions for Pom121, Nup37 and Nup35 and provide evidence for their copy numbers of 8, 32 and 16, respectively, per NPC. This approach can help pave the path for mapping the entirety of Nups in native NPCs and also other structural components of macromolecular complexes.


Subject(s)
Microscopy/methods , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Gene Dosage , HeLa Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...