Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Am Ann Deaf ; 168(5): 258-273, 2024.
Article in English | MEDLINE | ID: mdl-38766938

ABSTRACT

Little information is available on d/Deaf and hard of hearing (d/DHH) learners' L2 development. Their limited auditory access may discourage them from taking standardized tests, highlighting the need for alternative ways of assessing their L2 development and proficiency. Therefore, this study suggests adopting processability theory, which demonstrates a universal order of L2 development. Interviews with d/DHH learners and their teachers were conducted to explore their current difficulties in regard to understanding their L2 development. Also, we conducted brief speaking tasks to suggest alternatives to testing the L2 development of learners who are d/DHH in comparison to typical literacy learners. The result showed d/DHH students' L2 developmental patterns are similar to those of typical hearing peers, suggesting that d/DHH students and hearing learners share difficulties in similar areas when learning English. Teachers highlighted the lack of appropriate English tests to determine the d/DHH students' L2 development.


Subject(s)
Education of Hearing Disabled , Multilingualism , Humans , Education of Hearing Disabled/methods , Female , Male , Adolescent , Persons With Hearing Impairments/psychology , Students/psychology , Child , Language Tests , Deafness/psychology , Language Development , Comprehension
2.
Exp Mol Med ; 55(2): 283-289, 2023 02.
Article in English | MEDLINE | ID: mdl-36720916

ABSTRACT

Translation is mediated by precisely orchestrated sequential interactions among translation initiation components, mRNA, and ribosomes. Biochemical, structural, and genetic techniques have revealed the fundamental mechanism that determines what occurs and when, where and in what order. Most mRNAs are circularized via the eIF4E-eIF4G-PABP interaction, which stabilizes mRNAs and enhances translation by recycling ribosomes. However, studies using single-molecule fluorescence imaging have allowed for the visualization of complex data that opposes the traditional "functional circularization" theory. Here, we briefly introduce single-molecule techniques applied to studies on mRNA circularization and describe the results of in vitro and live-cell imaging. Finally, we discuss relevant insights and questions gained from single-molecule research related to translation.


Subject(s)
Poly(A)-Binding Proteins , Protein Biosynthesis , RNA, Messenger/metabolism , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Protein Binding , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism
3.
J Biol Chem ; 298(11): 102505, 2022 11.
Article in English | MEDLINE | ID: mdl-36126773

ABSTRACT

MutS homologs (MSHs) are highly conserved core components of DNA mismatch repair. Mismatch recognition provokes ATP-binding by MSH proteins that drives a conformational transition from a short-lived lesion-searching clamp to an extremely stable sliding clamp on the DNA. Here, we have expanded on previous bulk biochemical studies to examine the stability, lifetime, and kinetics of bacterial and human MSH sliding clamps on mismatched DNA using surface plasmon resonance and single-molecule analysis of fluorescently labeled proteins. We found that ATP-bound MSH complexes bound to blocked-end or very long mismatched DNAs were extremely stable over a range of ionic conditions. These observations underpinned the development of a high-throughput Förster resonance energy transfer system that specifically detects the formation of MSH sliding clamps on mismatched DNA. The Förster resonance energy transfer system is capable of distinguishing between HsMSH2-HsMSH3 and HsMSH2-HsMSH6 and appears suitable for chemical inhibitor screens. Taken together, our results provide additional insight into MSH sliding clamps as well as methods to distinguish their functions in mismatch repair.


Subject(s)
Escherichia coli Proteins , MutS DNA Mismatch-Binding Protein , Humans , Adenosine Triphosphate/metabolism , Base Pair Mismatch , DNA/metabolism , DNA Mismatch Repair , Escherichia coli Proteins/metabolism , MutS DNA Mismatch-Binding Protein/genetics , MutS DNA Mismatch-Binding Protein/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Proteins/genetics , Protein Binding
4.
Pharm Res ; 39(8): 1867-1880, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35778631

ABSTRACT

PURPOSE: To evaluate the duration of effect of rHuPH20 on SC absorption of cetuximab and to develop a mechanistic pharmacokinetic model linking the kinetics of rHuPH20 action with hyaluronan (HA) homeostasis and absorption of cetuximab from the SC space. METHODS: Serum pharmacokinetics of cetuximab was evaluated after IV and SC dosing at 0.4 and 10 mg/kg (control groups). In test groups, SC cetuximab was administered simultaneously with rHuPH20 (Co-Injection) or 12 h after injection of rHuPH20 (Pre-Injection). Mechanistic pharmacokinetic model was developed to simultaneously capture cetuximab kinetics in all groups. RESULTS: Administration of rHuPH20 resulted in a faster absorption of cetuximab; the difference between co-injection and pre-injection groups appeared to be dependent on the dose level. The model combined three major components: kinetics of rHuPH20 at SC site; HA homeostasis and its disruption by rHuPH20; and cetuximab systemic disposition and the effect of HA disruption on cetuximab SC absorption. The model provided good description of experimental data obtained in this study and collected previously. CONCLUSIONS: Proposed model can serve as a potential translational framework for capturing the effect of rHuPH20 across multiple preclinical species and in human studies and can be used for optimization of SC delivery of biotherapeutics.


Subject(s)
Hyaluronic Acid , Hyaluronoglucosaminidase , Animals , Cetuximab/pharmacology , Humans , Injections, Subcutaneous , Rats , Recombinant Proteins
5.
Biochem Biophys Res Commun ; 618: 73-78, 2022 08 27.
Article in English | MEDLINE | ID: mdl-35716598

ABSTRACT

Eukaryotic translation is a complex process that involves the interplay of various translation factors to convert genetic information into a specific amino acid chain. According to an elegant model of eukaryotic translation initiation, the 3' poly(A) tail of an mRNA, which is occupied by poly(A)-binding proteins (PABPs), communicates with the 5'-cap bound by eIF4E to enhance translation. Although the circularization of mRNA resulting from the communication is widely understood, it has yet to be directly observed. To explore mRNA circularization in translation, we analyzed the level of colocalization of eIF4E, eIF4G, and PABP on individual mRNAs in polysomal and subpolysomal fractions using single polysome analysis. Our results show that the three tested proteins barely coexist in mRNA in either polysomal or subpolysomal fractions, implying that the closed-loop structure generated by the communication between eIF4E, eIF4G, and PAPB may be transient during translation.


Subject(s)
Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Poly(A)-Binding Proteins/genetics , Polyribosomes/metabolism , Protein Binding , Protein Biosynthesis , RNA, Messenger/metabolism , Ribonucleoproteins
6.
Sci Adv ; 8(13): eabj3995, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35353579

ABSTRACT

Membrane nanotubes or tunneling nanotubes (TNTs) that connect cells have been recognized as a previously unidentified pathway for intercellular transport between distant cells. However, it is unknown how this delicate structure, which extends over tens of micrometers and remains robust for hours, is formed. Here, we found that a TNT develops from a double filopodial bridge (DFB) created by the physical contact of two filopodia through helical deformation of the DFB. The transition of a DFB to a close-ended TNT is most likely triggered by disruption of the adhesion of two filopodia by mechanical energy accumulated in a twisted DFB when one of the DFB ends is firmly attached through intercellular cadherin-cadherin interactions. These studies pinpoint the mechanistic questions about TNTs and elucidate a formation mechanism.

7.
Nucleic Acids Res ; 50(1): 306-321, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34904669

ABSTRACT

Poly(A)-binding protein (PABP) is a translation initiation factor that interacts with the poly(A) tail of mRNAs. PABP bound to poly(A) stimulates translation by interacting with the eukaryotic initiation factor 4G (eIF4G), which brings the 3' end of an mRNA close to its 5' m7G cap structure through consecutive interactions of the 3'-poly(A)-PABP-eIF4G-eIF4E-5' m7G cap. PABP is a highly abundant translation factor present in considerably larger quantities than mRNA and eIF4G in cells. However, it has not been elucidated how eIF4G, present in limited cellular concentrations, is not sequestered by mRNA-free PABP, present at high cellular concentrations, but associates with PABP complexed with the poly(A) tail of an mRNA. Here, we report that RNA-free PABPs dimerize with a head-to-head type configuration of PABP, which interferes in the interaction between PABP and eIF4G. We identified the domains of PABP responsible for PABP-PABP interaction. Poly(A) RNA was shown to convert the PABP-PABP complex into a poly(A)-PABP complex, with a head-to-tail-type configuration of PABP that facilitates the interaction between PABP and eIF4G. Lastly, we showed that the transition from the PABP dimer to the poly(A)-PABP complex is necessary for the translational activation function.


Subject(s)
Poly(A)-Binding Proteins/chemistry , Cell Line, Tumor , Eukaryotic Initiation Factor-4G/metabolism , Humans , Poly(A)-Binding Proteins/metabolism , Protein Binding , Protein Multimerization , RNA, Messenger/metabolism
8.
Autophagy ; 17(12): 4231-4248, 2021 12.
Article in English | MEDLINE | ID: mdl-33783327

ABSTRACT

Selective recognition and elimination of misfolded polypeptides are crucial for protein homeostasis. When the ubiquitin-proteasome system is impaired, misfolded polypeptides tend to form small cytosolic aggregates and are transported to the aggresome and eventually eliminated by the autophagy pathway. Despite the importance of this process, the regulation of aggresome formation remains poorly understood. Here, we identify TRIM28/TIF1ß/KAP1 (tripartite motif containing 28) as a negative regulator of aggresome formation. Direct interaction between TRIM28 and CTIF (cap binding complex dependent translation initiation factor) leads to inefficient aggresomal targeting of misfolded polypeptides. We also find that either treatment of cells with poly I:C or infection of the cells by influenza A viruses triggers the phosphorylation of TRIM28 at S473 in a way that depends on double-stranded RNA-activated protein kinase. The phosphorylation promotes association of TRIM28 with CTIF, inhibits aggresome formation, and consequently suppresses viral proliferation. Collectively, our data provide compelling evidence that TRIM28 is a negative regulator of aggresome formation.Abbreviations: BAG3: BCL2-associated athanogene 3; CTIF: CBC-dependent translation initiation factor; CED: CTIF-EEF1A1-DCTN1; DCTN1: dynactin subunit 1; EEF1A1: eukaryotic translation elongation factor 1 alpha 1; EIF2AK2: eukaryotic translation initiation factor 2 alpha kinase 2; HDAC6: histone deacetylase 6; IAV: influenza A virus; IP: immunoprecipitation; PLA: proximity ligation assay; polypeptidyl-puro: polypeptidyl-puromycin; qRT-PCR: quantitative reverse-transcription PCR; siRNA: small interfering RNA.


Subject(s)
Autophagy , Influenza A virus , Inclusion Bodies/metabolism , Influenza A virus/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
9.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619096

ABSTRACT

The pathogenic consequences of 369 unique human HsMLH1 missense variants has been hampered by the lack of a detailed function in mismatch repair (MMR). Here single-molecule images show that HsMSH2-HsMSH6 provides a platform for HsMLH1-HsPMS2 to form a stable sliding clamp on mismatched DNA. The mechanics of sliding clamp progression solves a significant operational puzzle in MMR and provides explicit predictions for the distribution of clinically relevant HsMLH1 missense mutations.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair , DNA-Binding Proteins/genetics , DNA/genetics , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Mutation, Missense , Binding Sites , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , DNA/chemistry , DNA/metabolism , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Models, Molecular , MutL Protein Homolog 1/chemistry , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/chemistry , MutS Homolog 2 Protein/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
10.
Nat Commun ; 11(1): 3106, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561765

ABSTRACT

Nonsense-mediated mRNA decay (NMD) typifies an mRNA surveillance pathway. Because NMD necessitates a translation event to recognize a premature termination codon on mRNAs, truncated misfolded polypeptides (NMD-polypeptides) could potentially be generated from NMD substrates as byproducts. Here, we show that when the ubiquitin-proteasome system is overwhelmed, various misfolded polypeptides including NMD-polypeptides accumulate in the aggresome: a perinuclear nonmembranous compartment eventually cleared by autophagy. Hyperphosphorylation of the key NMD factor UPF1 is required for selective targeting of the misfolded polypeptide aggregates toward the aggresome via the CTIF-eEF1A1-DCTN1 complex: the aggresome-targeting cellular machinery. Visualization at a single-particle level reveals that UPF1 increases the frequency and fidelity of movement of CTIF aggregates toward the aggresome. Furthermore, the apoptosis induced by proteotoxic stresses is suppressed by UPF1 hyperphosphorylation. Altogether, our data provide evidence that UPF1 functions in the regulation of a protein surveillance as well as an mRNA quality control.


Subject(s)
Nonsense Mediated mRNA Decay , Proteasome Endopeptidase Complex/metabolism , RNA Helicases/metabolism , RNA, Messenger/metabolism , Trans-Activators/metabolism , Unfolded Protein Response/genetics , Autophagy , Codon, Nonsense , Dynactin Complex/metabolism , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Imaging , Peptide Elongation Factor 1/metabolism , Phosphorylation , Protein Aggregates/genetics , Single Molecule Imaging , Ubiquitin/metabolism
11.
Nat Commun ; 10(1): 5294, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757945

ABSTRACT

A shared paradigm of mismatch repair (MMR) across biology depicts extensive exonuclease-driven strand-specific excision that begins at a distant single-stranded DNA (ssDNA) break and proceeds back past the mismatched nucleotides. Historical reconstitution studies concluded that Escherichia coli (Ec) MMR employed EcMutS, EcMutL, EcMutH, EcUvrD, EcSSB and one of four ssDNA exonucleases to accomplish excision. Recent single-molecule images demonstrated that EcMutS and EcMutL formed cascading sliding clamps on a mismatched DNA that together assisted EcMutH in introducing ssDNA breaks at distant newly replicated GATC sites. Here we visualize the complete strand-specific excision process and find that long-lived EcMutL sliding clamps capture EcUvrD helicase near the ssDNA break, significantly increasing its unwinding processivity. EcSSB modulates the EcMutL-EcUvrD unwinding dynamics, which is rarely accompanied by extensive ssDNA exonuclease digestion. Together these observations are consistent with an exonuclease-independent MMR strand excision mechanism that relies on EcMutL-EcUvrD helicase-driven displacement of ssDNA segments between adjacent EcMutH-GATC incisions.


Subject(s)
DNA Breaks, Single-Stranded , DNA Helicases/physiology , DNA Mismatch Repair/physiology , Escherichia coli Proteins/physiology , Escherichia coli/physiology , MutL Proteins/physiology , DNA Helicases/metabolism , DNA Repair/physiology , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Microscopy, Fluorescence , MutL Proteins/metabolism , Single Molecule Imaging
12.
BMB Rep ; 52(10): 589-594, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31401983

ABSTRACT

Single-molecule techniques have been used successfully to visualize real-time enzymatic activities, revealing transient complex properties and heterogeneity of various biological events. Especially, conventional force spectroscopy including optical tweezers and magnetic tweezers has been widely used to monitor change in DNA length by enzymes with high spatiotemporal resolutions of ∼nanometers and ∼milliseconds. However, DNA metabolism results from coordination of a number of components during the processes, requiring efficient monitoring of a complex of proteins catalyzing DNA substrates. In this min-review, we will introduce a simple and multiplexed single-molecule assay to detect DNA substrates catalyzed by enzymes with high-throughput data collection. We conclude with a perspective of possible directions that enhance capability of the assay to reveal complex biological events with higher resolution. [BMB Reports 2019; 52(10): 589-594].


Subject(s)
DNA/metabolism , Single Molecule Imaging/methods , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , High-Throughput Screening Assays , Optical Tweezers , Spectrum Analysis
13.
Nucleic Acids Res ; 47(18): e107, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31340015

ABSTRACT

Real-time visualization of single-proteins or -complexes on nucleic acid substrates is an essential tool for characterizing nucleic acid binding proteins. Here, we present a novel surface-condition independent and high-throughput single-molecule optical imaging platform called 'DNA skybridge'. The DNA skybridge is constructed in a 3D structure with 4 µm-high thin quartz barriers in a quartz slide. Each DNA end is attached to the top of the adjacent barrier, resulting in the extension and immobilization of DNA. In this 3D structure, the bottom surface is out-of-focus when the target molecules on the DNA are imaged. Moreover, the DNA skybridge itself creates a thin Gaussian light sheet beam parallel to the immobilized DNA. This dual property allows for imaging a single probe-tagged molecule moving on DNA while effectively suppressing interference with the surface and background signals from the surface.


Subject(s)
DNA/ultrastructure , High-Throughput Screening Assays/methods , Immobilized Nucleic Acids/ultrastructure , Single Molecule Imaging/methods , Nanotechnology/methods , Optical Imaging/methods
14.
Cognition ; 183: 269-276, 2019 02.
Article in English | MEDLINE | ID: mdl-30522041

ABSTRACT

This cross-modal priming study is one of the first to empirically test the long-held assumption that individual morphemes of multimorphemic words are represented according to a hierarchical structure. The results here support the psychological reality behind this assumption: Recognition of trimorphemic words (e.g., unkindness or [[un-[kind]]-ness]) was significantly facilitated by prior processing of their substrings when the substrings served as morphological constituents of the target words (e.g., unkind), but not when the substrings were not morphological constituents of the target words (e.g., kindness). This morphological structural priming occurred independently of the linear positions of morphological constituents.


Subject(s)
Pattern Recognition, Visual/physiology , Psycholinguistics , Reading , Recognition, Psychology/physiology , Speech Perception/physiology , Adult , Humans , Young Adult
15.
J Biol Chem ; 293(37): 14285-14294, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30072380

ABSTRACT

Sliding clamps on DNA consist of evolutionarily conserved enzymes that coordinate DNA replication, repair, and the cellular DNA damage response. MutS homolog (MSH) proteins initiate mismatch repair (MMR) by recognizing mispaired nucleotides and in the presence of ATP form stable sliding clamps that randomly diffuse along the DNA. The MSH sliding clamps subsequently load MutL homolog (MLH/PMS) proteins that form a second extremely stable sliding clamp, which together coordinate downstream MMR components with the excision-initiation site that may be hundreds to thousands of nucleotides distant from the mismatch. Specific or nonspecific binding of other proteins to the DNA between the mismatch and the distant excision-initiation site could conceivably obstruct the free diffusion of these MMR sliding clamps, inhibiting their ability to initiate repair. Here, we employed bulk biochemical analysis, single-molecule fluorescence imaging, and mathematical modeling to determine how sliding clamps might overcome such hindrances along the DNA. Using both bacterial and human MSH proteins, we found that increasing the number of MSH sliding clamps on a DNA decreased the association of the Escherichia coli transcriptional repressor LacI to its cognate promoter LacO. Our results suggest a simple mechanism whereby thermal diffusion of MSH sliding clamps along the DNA alters the association kinetics of other DNA-binding proteins over extended distances. These observations appear generally applicable to any stable sliding clamp that forms on DNA.


Subject(s)
DNA, Bacterial/metabolism , MutS DNA Mismatch-Binding Protein/metabolism , Thermus/metabolism , Adenosine Triphosphate/metabolism , Base Pair Mismatch , Models, Theoretical , Protein Binding , Surface Plasmon Resonance
16.
J Mol Biol ; 430(22): 4456-4468, 2018 10 26.
Article in English | MEDLINE | ID: mdl-29864444

ABSTRACT

DNA mismatch repair (MMR) is a DNA excision-resynthesis process that principally enhances replication fidelity. Highly conserved MutS (MSH) and MutL (MLH/PMS) homologs initiate MMR and in higher eukaryotes act as DNA damage sensors that can trigger apoptosis. MSH proteins recognize mismatched nucleotides, whereas the MLH/PMS proteins mediate multiple interactions associated with downstream MMR events including strand discrimination and strand-specific excision that are initiated at a significant distance from the mismatch. Remarkably, the biophysical functions of the MLH/PMS proteins have been elusive for decades. Here we consider recent observations that have helped to define the mechanics of MLH/PMS proteins and their role in choreographing MMR. We highlight the stochastic nature of DNA interactions that have been visualized by single-molecule analysis and the plasticity of protein complexes that employ thermal diffusion to complete the progressions of MMR.


Subject(s)
DNA/metabolism , MutL Proteins/metabolism , Single Molecule Imaging/methods , Animals , DNA Mismatch Repair , Humans , Kinetics , Signal Transduction , Stochastic Processes
17.
J Mol Biol ; 430(22): 4469-4480, 2018 10 26.
Article in English | MEDLINE | ID: mdl-29792877

ABSTRACT

DNA mismatch repair (MMR) corrects DNA base-pairing errors that occur during DNA replication. MMR catalyzes strand-specific DNA degradation and resynthesis by dynamic molecular coordination of sequential downstream pathways. The temporal and mechanistic order of molecular events is essential to insure interactions in MMR that occur over long distances on the DNA. Biophysical real-time studies of highly conserved components on mismatched DNA have shed light on the mechanics of MMR. Single-molecule imaging has visualized stochastically coordinated MMR interactions that are based on thermal fluctuation-driven motions. In this review, we describe the role of diffusivity and stochasticity in MMR beginning with mismatch recognition through strand-specific excision. We conclude with a perspective of the possible research directions that should solve the remaining questions in MMR.


Subject(s)
DNA/metabolism , Multiprotein Complexes/metabolism , Animals , Biophysical Phenomena , DNA Mismatch Repair , Diffusion , Humans , Stochastic Processes , Thermodynamics
18.
Cell Chem Biol ; 25(1): 57-66.e4, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29104063

ABSTRACT

The αɛθ core of Escherichia coli DNA polymerase III (Pol III) associates with the ß2 sliding clamp to processively synthesize DNA and remove misincorporated nucleotides. The α subunit is the polymerase while ɛ is the 3' to 5' proofreading exonuclease. In contrast to the polymerase activity of Pol III, dynamic features of proofreading are poorly understood. We used single-molecule assays to determine the excision rate and processivity of the ß2-associated Pol III core, and observed that both properties are enhanced by mutational strengthening of the interaction between ɛ and ß2. Thus, the ɛ-ß2 contact is maintained in both the synthesis and proofreading modes. Remarkably, single-molecule real-time fluorescence imaging revealed the dynamics of transfer of primer-template DNA between the polymerase and proofreading sites, showing that it does not involve breaking of the physical interaction between ɛ and ß2.


Subject(s)
DNA Polymerase III/metabolism , Escherichia coli/enzymology , Polymerization
19.
Opt Express ; 25(25): 32189-32197, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29245882

ABSTRACT

Real-time optical imaging combined with single-molecule manipulation broadens the horizons for acquiring information about the spatiotemporal localization and the mechanical details of target molecules. To obtain an optical signal outside the focal plane without unintended interruption of the force signal in single-molecule optical imaging-force spectroscopy, we developed an optical method to extend the depth of field in a high numerical aperture objective (≥ 1.2), required to visualize a single fluorophore. By axial scanning, using an electrically tunable lens with a fixed sample, we were successfully able to visualize the epidermal growth factor receptor (EGFR) moving along the three-dimensionally elongated filamentous actin bundles connecting cells (intercellular nanotube), while another EGFR on the intercellular nanotube was trapped by optical tweezers in living cells. Our approach is simple, fast and inexpensive, but it is powerful for imaging target molecules axially in single-molecule optical imaging-force spectroscopy.


Subject(s)
Actin Cytoskeleton/chemistry , ErbB Receptors/analysis , Lenses , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Optical Imaging/methods , Optical Tweezers , Spectrum Analysis/methods , HeLa Cells , Humans , Microscopy, Fluorescence/instrumentation , Nanotubes
20.
J Am Chem Soc ; 139(22): 7603-7615, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28493679

ABSTRACT

Photoswitching or modulation of quantum dots (QDs) can be promising for many fields that include display, memory, and super-resolution imaging. However, such modulations have mostly relied on photomodulations of conjugated molecules in QD vicinity, which typically require high power of high energy photons at UV. We report a visible light-induced facile modulation route for QD-dye conjugates. QD crystal violets conjugates (QD-CVs) were prepared and the crystal violet (CV) molecules on QD quenched the fluorescence efficiently. The fluorescence of QD-CVs showed a single cycle of emission burst as they go through three stages of (i) initially quenched "off" to (ii) photoactivated "on" as the result of chemical change of CVs induced by photoelectrons from QD and (iii) back to photodarkened "off" by radical-associated reactions. Multicolor on-demand photopatterning was demonstrated using QD-CV solid films. QD-CVs were introduced into cells, and excitation with visible light yielded photomodulation from "off" to "on" and "off" by nearly ten fold. Individual photoluminescence dynamics of QD-CVs was investigated using fluorescence correlation spectroscopy and single QD emission analysis, which revealed temporally stochastic photoactivations and photodarkenings. Exploiting the stochastic fluorescence burst of QD-CVs, simultaneous multicolor super-resolution localizations were demonstrated.

SELECTION OF CITATIONS
SEARCH DETAIL
...