Abstract

Our ability to detect, image, and quantify nanoscopic objects and molecules with visible light has undergone dramatic improvements over the past few decades. While fluorescence has historically been the go-to contrast mechanism for ultrasensitive light microscopy due to its superior background suppression and specificity, recent developments based on light scattering have reached single-molecule sensitivity. They also have the advantages of universal applicability and the ability to obtain information about the species of interest beyond its presence and location. Many of the recent advances are driven by novel approaches to illumination, detection, and background suppression, all aimed at isolating and maximizing the signal of interest. Here, we review these developments grouped according to the basic principles used, namely darkfield imaging, interferometric detection, and surface plasmon resonance microscopy.
1. Introduction
Structure and dynamics at the nanoscale determine the properties of matter and underpin the mechanisms of life. To understand and ultimately control these properties and functions, we must be able to visualize and thereby study matter on nanoscopic length scales. Much effort has thus been directed at the development and optimization of methods capable of detecting and characterizing nanoscopic objects. Broadly speaking, these can be divided into scanning probe approaches1−4 and those using the interaction between incident radiation and matter, including both electron microscopy and light microscopy. Light microscopy has been popular due to its noninvasive nature, dynamic operation, potential for high-throughput, and straightforward integration with other technologies. However, these capabilities come at the expense of inferior sensitivity and resolution, as compared to electron microscopy or scanning probe methods.
In the context of sensitivity, light microscopy arguably reached its ultimate limit more than 30 years ago with the detection of individual ions and molecules using absorption and fluorescence as contrast mechanisms.5−7 These single-molecule experiments were performed under cryogenic conditions with specifically selected chromophores to maximize the interaction strength between the incident light and the molecules of interest. Single-molecule experiments in the form of single-emitter fluorescence detection and imaging have since become routine.8 These experiments benefit from the specific advantage of fluorescence: the spectral separation between illumination and emission profiles, which enables efficient background suppression. This makes it possible to identify specific species in complex mixtures and optically noisy environments, such as living cells, as long as the surrounding objects are non- (or weakly) fluorescent at the combination of illumination and detection wavelengths. The ability to switch fluorophore emission on and off, both temporally and spatially, has also led to the emergence of super-resolution imaging techniques, in which the spatial resolution is now approaching the molecular scale.9−14
At the same time, using fluorescence as a contrast mechanism limits applicability of the most sensitive studies to species that are either highly fluorescent themselves or are made fluorescent by the introduction of external tags. Moreover, photochemical and photophysical processes limit both signal intensities and the degree to which signals are quantitative, which is often critical in the analytical context. These limitations, in combination with the prospect of a broader application scope, has driven the development of fluorescence-free methodologies. An alternative approach to separate illumination from detection is to exploit a geometric separation between them, as demonstrated by Siedentopf and Zsigmondy in 1902, who presented the ultramicroscope.15 The orthogonal illumination and detection pathways enabled the observation of light scattered by nanometer-sized gold colloids. Since this work, many different optical strategies have been developed to image scattered light in the absence of background, and they are grouped together under the term darkfield microscopy. Originating from Zernike’s phase contrast microscopy (PCM) in 1935, many technologies have used interference between transmitted (or reflected) incident and scattered light as the primary image contrast mechanism.16−18 These include differential interference contrast (DIC),19,20 interference reflection microscopy (IRM),21,22 and reflection interference contrast microscopy (RICM),23,24 all of which have predominantly focused on studies of tissues, cells, and supramolecular assemblies due to their lack of sensitivity for nano-objects sub-20 nm in diameter.
In fluorescence-free, extinction-based techniques, the contrast is generated from a combination of the scattering and absorption of incident light. Until recently, scattering-based techniques exhibited inferior sensitivity when compared to their absorption-based counterparts, which reached single-molecule sensitivity at room temperature a decade ago.25−27 Irrespective, the development of scattering-based techniques has continued toward the robust detection of single biological macromolecules, even in the absence of absorption. The main advantage of these techniques is their universal applicability, as any sample with a different refractive index to its surrounding medium will generate a signal. The key challenge in the context of studying nanoscale phenomena is thus to develop experimental configurations which are capable of detecting and isolating the smallest possible signals. This challenge is being addressed by a variety of efforts to reduce the imaging background or amplify the signal of interest.
In this review, we provide an overview of recent developments in approaches detecting light scattering with optical microscopes because of the underlying common architecture, although we would like to emphasize the remarkable levels of sensitivity achieved with alternative approaches, such as microcavities.28−30 After a brief introduction to the key concepts (section 2), we begin by reviewing advances in darkfield microscopy (section 3), focusing on approaches aimed at maximizing detection sensitivity by selecting photons that have interacted with the object of interest. A natural extension involves the use of illumination light to enhance the small signal that results from the weak interaction with the nano-object by interference (section 4). We conclude with an overview of efforts aimed at introducing additional signal amplification with plasmonically active substrates (section 5) before providing a brief summary and outlook (section 6).
2. Key Principles
The fundamental challenge in imaging on the nanoscale arises from the drop in interaction strength between the incident electromagnetic radiation and the object as the object becomes much smaller than the illumination wavelength, λ. This challenge can generally be compartmentalized into three key parameters: resolution, specificity, and sensitivity.
Spatial resolution, the minimum distance at which two distinct points in a specimen can be distinguished, can be divided into the lateral (X/Y) and axial (Z) dimensions. Resolution is limited by the diffraction of light as it passes through a circular aperture, such as an objective lens, ultimately restricting the achievable confinement of light and defining the imaged projection of a point-source, the point-spread function (PSF). This diffraction barrier was first introduced by Abbe,31 with the lateral resolution limited to λ/2NA, where NA is the numerical aperture of the objective lens, defined by the refractive index of the imaging medium multiplied by the sine of the maximum angle over which light can be gathered from the sample. Axial resolution is limited even further (2λ/NA2) due to the elliptical geometry of the PSF along this axis, creating an additional challenge for resolving objects in three dimensions.
In the context of microscopy, specificity refers to the ability to distinguish signals from different types of objects of interest within a sample. This can be achieved by a variety of strategies, including: analysis of the magnitude of a detected signal, surface functionalization to promote specific binding and attachment of multiple different fluorescence or scattering labels. For the techniques discussed in this review, specificity relies on these strategies as chemical specificity is not inherently encoded within Rayleigh scattering (in contrast to Raman scattering).33,34
Finally, sensitivity refers to the ability of an instrument to not only detect a signal from an object but to differentiate it from any unwanted background signal, or noise. For an ideal experiment, with no external or environmental noise, the noise-floor is limited by the properties of the detector, such as dark noise and read noise, as well as the quantum noise (referred to as shot noise) associated with the counting of discrete particles (photons) arriving at the detector. The width of this Poissonian intensity distribution thus scales with √N, where N is the number of photons detected. Because of the well-defined geometry of the PSF described above, it is possible to localize its center of mass with subdiffraction limited precision by fitting the PSF to a 2D Gaussian function,35 which enables high-resolution tracking of the motion of an object of interest. Localization precision is limited by the number of photons detected and can be approximated as σ/√N in a shot-noise-limited experiment, where σ is the standard deviation of the PSF fit.36 This leads to an interplay between the localization precision and temporal resolution of a tracking experiment due to the limited number of photons that can be detected per unit time. In fluorescence-based approaches, the number of photons detected per unit time is limited by optical saturation of the fluorophore. In scattering-based approaches, there is no a priori limited photon budget provided by the scattering object, so the limitation is determined by either sample/optical damage thresholds or saturation of the detector.
Intuitively, the strength of the interaction between light and an object should scale with the polarizability of the object, which is a measure of its ability to respond to and interact with an incident electromagnetic wave. Assuming that the object of interest can be approximated as a sphere, which is often a good approximation for nanoscopic objects much smaller than the diffraction limit, it can be treated as a point dipole with a polarizability given by
| 1 |
where εm/p is the complex permittivity of the medium and particle respectively, and V is its volume.37 The polarizability then yields the scattering and absorption cross sections as
| 2 |
| 3 |
which sum to yield the extinction cross-section, σext. For nonresonant particles, σscat dominates the detected signal. The dependence of σscat on the square of the volume of the particle is primarily responsible for the difficulty of detecting very small objects without the use of a label: a reduction in the diameter of a sphere by a factor of 10 reduces the intensity of scattered light by 106 for a given incident photon flux. While this can be somewhat alleviated by plasmonic scattering enhancement for materials such as silver and gold, the volume of the nanoparticle still dominates the magnitude of its scattering signal.
Generally speaking, when shining light on a sample, the detected signal intensity at a given point in the image (Idet) can be written as the sum of the fields associated with the relatively unperturbed incident light (Ei) and the light scattered by the presence of an object (Es):
| 4 |
The scattered light field is proportional to the complex polarizability of the object, implying both linear and quadratic dependencies of the two signal contributions on the volume of the object, a feature that can be taken advantage of. In the context of extinction measurements, the cross-term in eq 4 indicates that spatial mode matching between the wavefronts of the two fields is desirable for efficient extinction to occur. A tightly focused Gaussian beam has a stronger dipolar character compared to plane wave illumination and hence improves the interaction with a dipolar nanoscopic scatterer resulting in increased extinction efficiency.38 This effect on the extinction signal is somewhat unsurprising if considered from the perspective of maximizing the ratio of the extinction cross-section to the area of the illumination profile (σ/A).
It is also important to note that, for a point source located near a refractive index interface, a high proportion of the incident light is scattered into the higher index material, resulting in this light being collected at large angles by the objective lens.39−41 Consequently, illuminating the sample via the low NA regions of the objective, such as in wide-field illumination, provides an opportunity to spatially discriminate between the scattered and transmitted/reflected illumination light in the back focal plane (BFP) of the objective. This can be advantageous when attempting to manipulate eq 4, and therefore the contrast of the final image, by perturbing one field relative to the other.42−45
Thus far, we have only considered the signal arising from the object of interest, but the presence of background light poses the most significant limitation to object sensitivity. Unwanted background light can emanate from multiple sources, such as other scattering objects in the sample, surface inhomogeneity of the sample substrate (typically a glass coverslip), or stray reflections from components in the optical system. For conventional brightfield illumination, the largest background contribution arises from the illumination field, which renders the small signal from a nano-object signal very difficult to detect.
All approaches discussed in this review attempt to manipulate the relative magnitudes of the different contributions in eq 4, with the ultimate goal of detecting and in some cases obtaining additional information about the object through Es. Darkfield imaging (section 3) refers to methods that minimize any light that has not interacted with the object of interest, Ei, in detection. The focus is thus on detecting and quantifying |Es|2, which becomes increasingly challenging the smaller the object becomes due to the V2 scaling in the presence of competing background signals. Alternatively, one can attempt to reduce the relative magnitude of background signals by focusing on the interference term 2Re(EiEs*), which is the target of interferometric microscopy (section 4). As both of these methods will eventually face the limitation of finite illumination powers and polarizabilities, additional steps can be taken to increase Ei beyond what can be achieved in a straightforward illumination approach, for instance by taking advantage of plasmonic effects (section 5).
3. Darkfield Imaging
3.1. Introduction
The most intuitive way to maximize the sensitivity of a microscope is to isolate the photons that interacted with the species of interest from any background signal, a concept optimized in single molecule fluorescence imaging, where this isolation is achieved by filtering for photons of specific wavelengths during both illumination and detection. In the context of scattering detection, analogous efforts to maximize background rejection can be broadly grouped under the term darkfield microscopy.46−50 The major difference, in this sense, between fluorescence imaging and darkfield microscopy is that the illumination light is of the same wavelength as the scattered light, meaning that background rejection cannot be achieved by orthogonal optical filters. In terms of eq 4, we eliminate Ei, as far as possible, meaning that in an ideal scenario:
| 5 |
In practice, most approaches are variations on the original theme introduced by Siedentopf and Zsigmondy more than a century ago.15 While field separation in this case was achieved by detecting scattered light orthogonally to the direction of illumination, this has since been achieved through the use of appropriately placed beam stops, apertures, or reflective optics. In the context of this review, the key limitation and challenge faced by darkfield approaches, in the spirit of detecting ever smaller objects, can be traced to the V2 scaling of the purely scattered term and the presence of imaging background. As a result, darkfield microscopes perform well when detecting and imaging objects with a diameter on the order of >20 nm but struggle immensely to reach the 5 nm scale required for single macromolecules or small nanoparticles. While light scattered by such small objects could still in principle be detected, this rapid drop in scattered power has in practice been overwhelmed by stray light from the sample or any components in the imaging setup. Even if the background signal intensity were fixed over the course of a measurement, such that it could be isolated from the analyte signal and thus subtracted from the final image, an extremely high dynamic range measurement would be required to detect the signal from a single molecule. Most recent efforts aimed at improving the performance of darkfield microscopes have thus focused on innovative approaches to separate the scattered light from the background light, and in this section we review those approaches.
3.2. Total Internal Reflection Scattering
Total internal reflection (TIR) refers to the reflection of incident light at a refractive index boundary and provides a convenient means of containing the illumination light at the glass–water interface. This is usually achieved by directing a laser beam onto the sample substrate at an angle above the critical angle, which generates an evanescent field within a few hundred nanometers of the surface of the substrate. This type of illumination has been heavily utilized in total internal reflection fluorescence (TIRF) microscopy.51−53 TIR illumination can be advantageous in darkfield microscopy as the signal ideally only contains contributions from scatterers close to the surface of the substrate, thereby minimizing stray light. The first use of TIR illumination in a darkfield microscope was presented by Ambrose in 1956, where cells in water were imaged on the surface of a glass slide, which was illuminated above the critical angle via a glass prism (Figure 1A).54,55 In this section, we will focus on objective-type total internal reflection scattering (TIRS) microscopy, which was introduced as an alternative to prism-based illumination by Braslavsky et al.56 In objective-type TIR illumination, the critical angle is reached by using the outer edges of the entrance pupil of a suitably high-NA oil immersion objective, as previously demonstrated by Stout and Axelrod in TIRF microscopy.57 In the original objective-type TIRS study,56 two different optical set-ups were suggested, both based on an inverted microscope and using a beamsplitter to direct the illumination into the objective lens and a field stop to block the back-reflected beam at a position where the BFP of the objective lens is reimaged. The authors demonstrated the sensitivity of the technique by imaging 20 nm gold nanoparticles (GNPs) and 60 nm polystyrene beads.
Figure 1.
Commonly employed approaches to scattering-based total internal reflection microscopy. (A) Prism-type total internal reflection microscopy. (B) Objective-type total internal reflection microscopy. P, prism; OBJ, objective; L, tube lens; M, mirror; BB, beam block; C, camera. (C) Image of a 20 nm GNP on a glass coverslip taken with an optimized objective-type TIRS microscope, scale bar = 0.5 μm. (D) Schematic illustrating the fields involved with TIRS microscopy, Ei incident field, Es scattered field, Ee evanescent field, θ incident angle of illumination, which is greater than the critical angle, θc. (C) adapted with permission from ref (58). Copyright 2021 Philipp Kukura.
While we refer throughout this section to reported performance characteristics, such as localization precision and temporal resolution, these parameters are not necessarily comparable across techniques. The reason is that the ultimate performance depends on the details of the experimental arrangement beyond the optical approach: detection efficiency, mechanical stability, illumination power, and alignment details in terms of background suppression. Nevertheless, the reported literature values provide an indication of what is possible and has been demonstrated, which should be considered in combination with the approach as a whole.
For objective-type TIR, the key challenge involves coupling the illumination light in and out of the imaging objective, while minimizing the amount of stray light reaching the detector. Generally, this is best achieved by placing the reflective elements as close as geometrically possible to the entrance pupil of the objective, which minimizes back reflections from the multiple lenses within the objective (Figure 1B). While reimaging is a possibility, as in the original study by Braslavsky et al.,56 focusing a laser beam into the BFP of a high-NA objective lens produces a number of divergent reflections, which can be difficult to eliminate in a reimaging configuration. In objective-based darkfield techniques, there is an interplay between background suppression and the effective NA of the optical system, as blocking stray light reduces scattered light collection.
Noji and co-workers approached this problem by introducing a perforated mirror.59 The concept behind perforated mirror TIR darkfield is to sacrifice spatial resolution and collection efficiency in exchange for background suppression. The optical setup is based on a standard TIRF microscope, with the dichroic mirror exchanged for a perforated mirror, a mirror with an elliptical hole in the center that allows a portion of the scattered light to pass while blocking the reflected illumination beam (Figure 2A). When viewed along the optical axis, the elliptical hole in the perforated mirror acts as a circular aperture, which prevents the introduction of aberrations from asymmetrically cutting the NA (Figure 2B). In proof-of-principle experiments, 40 nm GNPs were imaged with 0.1 ms temporal resolution on a glass surface (Figure 2C). The resulting SNR was 1.5–2.1 times higher for perforated mirror TIR darkfield as compared to an epi-illumination darkfield microscope with a dot mirror. This improvement resulted from the increase in background suppression and occurred despite a considerable drop in the NA of the optical system caused by the interception of scattered light by the perforated mirror.
Figure 2.
Perforated mirror TIR darkfield microscopy. (A) Schematic demonstrating the separation of scattered light from the reflected illumination beam. OBJ, objective lens; PM, perforated mirror. (B) View from the back aperture of the objective looking down the optical axis. (C) Image of 40 nm GNP in water immobilized on a glass slide. (D) Rotational motion of 40 nm gold-nanoparticle-labeled F1-ATPase at 2 mM ATP, with the inset displaying an example trace from which the data is obtained. (C,D) Adapted with permission from ref (59). Copyright 2010 Elsevier.
In fluorescence-free microscopy, GNPs are regularly used as scattering labels due to the relatively large scattering cross-section for the particle size as a result of plasmonic enhancement. This concept was combined with perforated mirror TIR darkfield to study multiple molecular motors with high spatiotemporal resolution. Labeling the γ-subunit of the F1-ATPase motor with a 40 nm GNP enabled the rotational motion of the motor to be tracked with 9.1 μs time resolution and a localization precision on the order of 1–2 nm (Figure 2D). Building on this work, the rotational dynamics of the F1-ATPase,60−64 V1-ATPase,65,66 and V0V1-ATPase67 motors have been studied using this technique. A similar labeling approach was used to study the stepping mechanism of the myosin VI motor, obtaining a spatiotemporal resolution of 2 nm and 37 μs.68 Isojima et al. used the technique to visualize intermediate states between the 8 nm steps of the kinesin-1 motor by probing the motion of a kinesin head with a temporal resolution of 55 μs and a spatial precision of 1.3 nm.69 In addition, Nakamura et al. studied the ∼50 nm/s stepping movement of Settatia marcescens chitinase A and detected the 1 nm forward and backward steps this motion consists of by tracking the position of the scattering label with 0.3 nm localization precision and 0.5 ms temporal resolution.70
In addition to studies using scattering labels like those highlighted above, perforated mirror TIR darkfield microscopy has been used for label-free detection of ∼100 nm diameter influenza virus particles, immobilized on glass with a detection limit of 1.2 × 104 plaque-forming units (PFU)/mL.71 The authors used scanning electron microscopy (SEM) and fluorescence microscopy to confirm that the darkfield microscope was capable of imaging single influenza virus particles as opposed to aggregates.
Recently, perforated mirror TIR darkfield microscopy has undergone multiple technological developments. Iino and co-workers introduced an axicon lens into the illumination path to generate an annular illumination profile at the BFP of the objective.72 This method enabled greater illumination intensities compared to previous implementations, as the illumination intensity was limited by the damage threshold of the objective lens when irradiated with a stationary, focused beam. This illumination scheme improved the localization precision of the technique to 1.3 Å for 40 nm GNPs and 1.9 Å for 30 nm GNPs with 1 ms time resolution. This development was applied to study the stepping motion of processive dynein with 100 μs temporal resolution and 0.7 nm localization precision.73 A further development by the same group introduced a multiple-wavelength illumination scheme, which is capable of tracking different types of metal nanoparticles simultaneously by exploiting their associated plasmon resonances and introducing a spectrometer in the imaging path.74
An alternative strategy for separating scattered from background light when using TIR illumination involves introducing a pair of micromirrors close to the back aperture of the microscope objective to couple illumination in and out of the imaging path. This arrangement can yield very high optical efficiency, with 94% of the collected backscattered light passing the micromirrors.58 In addition, the approach is capable of highly effective background suppression through the interception of back reflections from the objective. The sensitivity of the technique was demonstrated by imaging 20 nm GNPs in water immobilized on a glass coverslip with shot-noise-limited localization precision and a signal-to-background ratio close to 100. As a proof of principle, the authors tracked the position of 20 nm silver and GNPs diffusing at an oil–water interface with ∼1 nm spatial precision and 10 μs temporal resolution, using stroboscopic illumination.
In addition to tracking the location of the scattering label, polarization-sensitive darkfield microscopy is capable of simultaneously determining the orientation of gold nanorods (GNRs)75 due to the strongly polarized scattering response of the nanoparticle.37,76 In a recent demonstration of this concept, Mazaheri et al. tracked 71 nm × 25 nm GNRs diffusing on a supported lipid bilayer with a temporal resolution of 2.3 μs, lateral localization precision of 3 nm, and rotational precision of 4°, using a polarization-sensitive objective-type TIRS microscope.77
3.3. Rotating Coherent Scattering Microscopy
As noted by Abbe in 1873, oblique, coherent illumination of a sample results in a factor of 2 improvement to the achievable spatial resolution when compared to illumination at normal incidence,31,78 due to higher orders of diffraction being collected by the objective lens. Illuminating two adjacent objects at an oblique angle and in the direction of the alignment of the two objects results in a time-delay between the illumination of each object. For coherent illumination, this induces a phase-offset between the scattered fields emanating from the two objects, which interfere at the detector (Figure 3C). Destructive interference between the scattered waves allows an intensity separation of the two objects at a distance of ∼λ/4 apart. Many structured coherent illumination and synthetic aperture imaging techniques have demonstrated improved resolution by leveraging this effect.79−83
Figure 3.
ROCS microscopy principle and representative results. (A) Schematic of ROCS microscopy. OBJ, objective; BS, beamsplitter. (B) Translation of the illumination beam in a ring pattern around the back aperture of the objective lens. (C) Schematic of interference between adjacent scatterers <λ/2 apart. Under the condition of normal incidence, the scattered waves constructively interfere at the detector, whereas for oblique illumination, a phase delay of Δφ = π is introduced between the two scattered waves, leading to destructive interference at the detector and enabling the scatterers to be resolved. (D) TIR-darkfield ROCS image of 200 nm polystyrene beads illumination from a single azimuthal angle. (E) An image from the same sample once the beam is scanned in the pattern from B during a single detector exposure. (F) and (G) are the TIR-brightfield equivalents to D and E, respectively. (C) adapted with permission from ref (85). Copyright 2016 Springer Nature under Creative Commons Attribution 4.0 International License https://creativecommons.org/licenses/by/4.0/. (D–G) adapted with permission from ref (91). Copyright 2018 Optical Society of America.
When illuminating complex samples containing multiple scattering objects in this way, however, the interference of coherent scattering events produces unwanted speckle patterns and interference artifacts. One approach of overcoming these artifacts is to illuminate the sample sequentially from all directions. In this way, for any orientation of adjacent objects, there will be two instances where they are illuminated from opposite directions along their alignment axis. Incoherent addition of the coherent images results in a suppression of the coherence artifacts and a strong separation contrast. This concept is the basis for rotating coherent scattering (ROCS) microscopy, which was first introduced by Rohrbach and co-workers.84−88
In the first iteration of TIR darkfield ROCS microscopy, a spatial light modulator was used to deflect the illumination beam to acquire 72 partial images, each taken with TIR illumination at a different azimuthal angle, which were incoherently averaged to obtain a final image.84 In this experimental setup, the scattered light was separated from background light by placing a field stop in a plane conjugate to the objective’s BFP. In proof-of-principle experiments, 190 nm polystyrene beads were imaged in water to showcase the improved spatial resolution. A potential downside of ROCS microscopy is that minor image distortions are introduced,78 with scattering objects appearing to be located further apart than their physical distance.
Introducing a two-axis scan mirror, which scanned the beam in a ring pattern across the BFP of the objective within a single exposure of the detector (Figure 3A,B),85 removed the need for post processing and enabled an acquisition rate up to 100 Hz. The technique was applied to image J774 mouse macrophages and 200 nm thin helical bacteria with a frame rate of 63 and 100 Hz, respectively, achieving spatial resolution of ∼150 nm despite the decrease in effective NA of the optical system due to the field stop blocking the reflected illumination beam in the detection path. The authors demonstrated the improvement in spatial resolution, by comparing TIR-ROCS images with TIRF images acquired using the same objective lens and a similar detection wavelength, which yielded a spatial resolution of ∼210 nm. Alternatively, darkfield ROCS can be performed without relying on TIR, which can be advantageous when studying species which are located away from the surface of the coverslip.89 The authors used this approach in conjunction with optical tweezers and interferometric BFP tracking to study the bending of microtubules attached to optically trapped beads.
In addition to operating in darkfield mode, ROCS can be operated in brightfield mode by removing the darkfield stop, enabling interference between the collected scattered light and the back-reflected light. Brightfield ROCS was introduced by Jünger and Rohrbach, who used it in combination with TIR-darkfield ROCS to investigate the activity of the cytoskeleton of J774 mouse macrophages.90 This approach was further developed by utilizing the full collection efficiency of a 1.46 NA objective lens, leading to higher detection NA at the expense of lower imaging contrast, relative to an identical setup with a darkfield stop in the detection path.91 This was demonstrated by imaging 200 nm polystyrene beads with both imaging modalities (Figure 3D–G). In both darkfield and brightfield ROCS, it is clear that the coherence artifacts present in images acquired with illumination from a single oblique angle are not present in images acquired with the illumination averaged over all azimuthal angles. By controlling the spatial coherence of the illumination light with a rotating diffuser, the authors showed that maximizing the spatial coherence of the illumination light leads to optimum image contrast and resolution.
3.4. Objective Coupling Mirror
In addition to operating with TIR illumination described above, efficient separation of illumination from detection can be achieved in epi-illumination by a single, small mirror which directs the illumination into the objective lens and allows the returning scattered light to pass. This concept was implemented by Xie and co-workers, who used a small circular mirror attached to a glass window to couple the illumination beam diagonally into the objective.92 The small mirror enabled the majority of scattered and reflected light to pass, with separation obtained by inserting a field stop after the first lens in the detection path, due to the offset locations of the scattered and reflected light image planes. The authors used this microscope to track 100–150 nm GNPs endocytosed within lysosomal vesicles inside live cells, moving along microtubules and being transported by molecular motors, with 25 μs temporal resolution and 1–2 nm spatial precision using a quadrant photodiode (QPD) in detection. The diagonal illumination approach used in this microscope was adapted from the scheme developed by Nishiyama et al., who combined transmission darkfield microscopy with an optical trap and demonstrated the presence of ∼4 nm substeps within the 8 nm step of kinesin.48 The ∼4 nm substeps elucidated in this study were recently observed using multiple different techniques.93,94
An alternative version of this approach was developed by Berry and co-workers. In their version, a small rod mirror situated close to the back aperture of the objective lens directs the illumination along the optical axis (Figure 4A).95 The rod mirror and its mount blocked less than 10% of the area of the back aperture of the objective lens while intercepting back reflections from the objective and interfaces within the sample due to its position (Figure 4B). They tested the microscope performance by imaging a 100 nm GNP bound to a flagellar motor on top of an Escherichia coli cell (Figure 4C). The low background signal from the Escherichia coli cell, relative to the signal generated by the 100 nm GNP, was attributed to the poor backscattering efficiency of the cell, as the cell is a significantly larger particle which scatters the majority of the light in the forward direction - a potential benefit for further in vivo applications. As a proof-of-principle demonstration, the microscope was also used to track the rotation of the bacterial flagellar motor at a frame rate of 110 kHz by labeling the hook of the motor with a 100 nm GNP (Figure 4D). Further in vivo studies of that motor using the same technique generated torque-speed curves of the motor under various conditions and allowed measurement of stepwise speed changes at low load.96,97
Figure 4.
Schematic and data from an objective coupling mirror darkfield microscope. (A) Schematic of the experimental setup. OBJ, objective lens; RM, rod mirror. (B) View from the back aperture of the objective looking down the optical axis toward the rod mirror. (C) Image of a 100 nm GNP attached to a flagellar motor on top of an Escherichia coli cell, scale bar = 1 μm. (D) Traces of the displacement of a 100 nm GNP attached to a bacterial flagellar motor, the black and gray lines correspond to X and Y displacement, respectively. (C,D) Adapted with permission from ref (95). Copyright 2010 AIP Publishing.
As a further demonstration of epi-illumination objective coupling mirror darkfield, Noji and co-workers employed polarization-sensitive detection75 and defocused image analysis98 to track both the position and orientation of 80 nm × 40 nm GNRs, with 10 μs temporal resolution and ∼1° azimuthal angle precision.99 In addition to determining the azimuthal angle, the defocused image analysis method was capable of simultaneously extracting the polar angle of the GNRs with ∼1° precision.
3.5. Darkfield Stop
Another strategy to obtain efficient separation of background and scattered light combines a field stop with transmission illumination (Figure 5A).42 This strategy leads to highly efficient background suppression (>107) because the illumination beam does not pass through the objective toward the sample, significantly reducing reflections being directed toward the camera. In the setup devised by Weigel et al., the darkfield stop was fabricated from blackened aluminum and was significantly larger than the illumination beam, sacrificing scattered light collection efficiency for superior background suppression (Figure 5B). The sensitivity of the microscope was demonstrated by imaging 20 nm GNPs in water immobilized on a glass coverslip (Figure 5C) and 10 nm GNPs binding to a glass coverslip in water at an acquisition rate of 100 Hz. The high background suppression enabled single molecule fluorescence detection without the use of an optical filter (Figure 5D,E).
Figure 5.
Darkfield stop schematics and performance. (A) Schematic illustrating the separation of illumination and scattered light. OBJ, objective; DS, darkfield stop. (B) View from the back aperture of the objective lens looking along the optical axis toward a 4 mm darkfield stop. (C) Image of 20 nm GNPs in water with 1 ms integration time. (D) Fluorescence image of ATTO 610 molecules without the use of an optical filter with 100 ms integration time. (E) Traces of the fluorescence emission of the molecules labeled in (D). (C–E) adapted with permission from ref (42). Copyright 2014 American Chemical Society.
3.6. Darkfield Imaging Using Optical Waveguides
In addition to objective-based darkfield microscopy, the sample can also be illuminated by an evanescent field generated as the light propagates through a waveguide. In waveguide scattering microscopy (WSM), the scattered light is efficiently separated from the background by aligning the illumination and detection pathways orthogonal to each other (Figure 6A,D). Evanescent illumination using waveguides has also been utilized in fluorescence microscopy.100−102
Figure 6.
Darkfield imaging in optical waveguides. (A–C) Evanescent scattering using waveguide illumination. (A) Schematic of the experimental setup. OBJ, objective; SMF, single mode fiber; WC, waveguide core. (B) Image of 150 nm fluorescently labeled lipid vesicles, scale bar = 2 μm. (C) Scattering intensity trace of 18 nm GNPs binding to a 100 nm lipid vesicle. (D–F) Darkfield imaging in a nanofluidic optical fiber. (D) Schematic of experimental design. OBJ, objective; WC, waveguide core; NC, nanochannel. (E) Example traces of CCMV virions diffusing in the optical fiber. (F) Image of a mixture of 19–51 nm polystyrene beads. (B,C) adapted with permission from ref (106). Copyright 2015 American Chemical Society. (E,F) adapted with permission from ref (112). Copyright 2015 American Chemical Society.
WSM was first introduced by Thoma et al. in 1997, who applied the technique to image thin films using an evanescent field penetrating the surface of an ion-exchanged slab waveguide.103 One advantage of waveguide-based evanescent illumination over objective-based evanescent illumination is that the depth of the evanescent field can be tailored over a much larger range by adapting the geometry and refractive indices of the materials used to construct the waveguide.101 In addition, the orthogonal placement of the objective lens relative to the guided wave removes the need for placing reflective optics, apertures, or beam blocks in the detection path, and a much larger field of view (FOV) can be illuminated. Ni et al. applied the technique to nanoparticles by imaging GNRs as small as 85 ± 9 × 30 ± 4 nm, coated on the core surface of an etched multimode optical fiber.104 Cahoon and co-workers built on this concept by imaging ∼150 nm × 50 nm GNRs and silicon nanowires with diameters down to 50 nm, using glass coverslips and microscope slides as waveguides.105 In this study, the authors presented different experimental configurations capable of detecting the forward and backscattered light from the nanoparticles.
Agnarsson et al. introduced a waveguide chip that was capable of single particle fluorescence and scattering measurements (Figure 6A).106,107 In this technique, the sample was exposed to the core of the waveguide by etching a sample well into the polymer cladding layer on top of the silica core (Figure 6A). Light was coupled in to the single-mode waveguide via a single-mode fiber, which generated an evanescent field that penetrated the core–water interface. Particles binding to the core–water interface were detected through scattering of the evanescent field, which was collected by a water immersion objective lens oriented perpendicular to the waveguide. To generate a low-intensity background, the waveguide chip was constructed from an extremely smooth silica core sandwiched between two layers of CYTOP, a fluorinated polymer with a refractive index of 1.34, close to that of water. As a proof-of-concept experiment, the chip was used to image fluorescently labeled lipid vesicles with a diameter of 150 nm, demonstrating that the chip was capable of detecting the light scattered by biological nanoparticles (Figure 6B). Furthermore, the technique was used to analyze the kinetics of phospholipase A2 enzymes digesting 100 nm vesicles and to image individual 18 nm GNPs binding to lipid vesicles with a diameter of 100 nm (Figure 6C). Building on this initial work, the technique was applied to analyze the size-dependent binding of ligand-functionalized GNPs to lipid membranes.108 In other studies, the chip’s ability to enable imaging of single vesicles using both scattering and fluorescence has been used to study the formation of lipid membranes,109 the lipid content of extracellular vesicles,110 and the effect of the addition of monomeric α-synuclein on various types of lipid vesicles.111
In addition to utilizing the evanescent field that is generated as the light propagates through the waveguide, it is also possible to illuminate with the guided laser mode directly. Manoharan and co-workers designed a single-mode silica fiber containing a subwavelength cylindrical nanofluidic channel to image nanoparticles with high levels of sensitivity (Figure 6D).112 The nanoparticles were illuminated with a guided optical mode that was coupled into the fiber using a low-magnification objective lens. A portion of the light scattered by the nanoparticles was collected by an oil immersion objective, which was oriented perpendicular to the illumination. The fiber exhibited very low background scattering, leading to a low-intensity, stationary background which was removed computationally. In proof-of-principle experiments, a mixture of polystyrene beads ranging from 19 to 51 nm traveling through the fiber were imaged and tracked with a frame rate of 1 kHz (Figure 6F). The localization error of the technique ranges from <50 to 300 nm depending on the size of the nanoparticle, with the error largely induced by motion blur at the limited frame rate of the detector. In addition, the technique was used successfully to image and track wild-type cowpea chlorotic mottle virus (CCMV) virions with a diameter of 26 nm (Figure 6E), and was capable of operating with frame rates of up to 3.5 kHz. Building on this work, Faez et al. developed a technique termed nanocapillary electrophoretic tracking (nanoCET), which combined the technique described above with two microelectrodes, both oriented perpendicular to the fiber and situated along its plane.113 An alternating voltage was applied between the two electrodes to drive a 60 nm GNP back and forth along the channel at a frequency of up to 200 Hz, leading to measurements of the electrophoretic mobility of the particle at a frame rate of 2 kHz.
Jiang et al. advanced fiber-based scattering microscopy by introducing an optical fiber capable of tracking nanoparticles in three dimensions.114 The microfluidic channel was displaced from the core of the fiber in one dimension so that a homogenized evanescent field was generated in this dimension of the channel. This enabled the 3D position of the particle to be extracted by analyzing the location and intensity of the PSF. Proof-of-principle experiments tracked 50 nm GNPs in three dimensions at a frame rate of 2 kHz, with <3 nm and ∼21 nm localization accuracy in the lateral and axial dimensions, respectively. In this initial study, to obtain the evanescent illumination conditions described above, the refractive index of the liquid in the microchannel was set to 1.44. Improvement of the data analysis procedure extended this method to more complex illumination profiles, enabling the channel to be filled with water and paving the way to biological studies.115 Another technological development was introduced by Förster et al., who incorporated a hollow-core fiber with a channel diameter over an order of magnitude larger than that used by Manoharan and co-workers.112,116,699 This fiber enabled more particles to be studied simultaneously. In initial experiments, 40 nm GNPs andEscherichia coli virus λ phages were imaged and tracked. In those experiments, the sensitivity was lower than in aforementioned experiments conducted by Manoharan and co-workers due to the incorporation of a low-magnification, low-NA oil immersion objective, as resolution and the collection efficiency were sacrificed in exchange for a large depth of field.
4. Interferometric Imaging
4.1. Introduction
Since the introduction of PCM in 1935,16 many optical microscopy technologies have employed interference as the contrast mechanism. Despite the developments introduced by DIC microscopy,19,20 IRM,21 and RICM,23 ultrasensitive optical detection of nonfluorescent nano-objects remained largely limited to darkfield microscopy. As discussed in the previous section, however, the square-dependence of the optical signal intensity on object volume in darkfield microscopy has resulted in real-world limitations to the achievable detection sensitivity. These limitations can be circumvented to some degree by the introduction of a reference light field to the optical signal. The resulting intensity at the detector, Id, then becomes a superposition of the reference field, Er = Ereiϕr, and the field scattered from the probed nano-object, Es = Eseiϕs, such that eq 4 now reads:
| 6 |
where Ir and Is represent the intensity of the reference and scattered light respectively, and ϕ = ϕr – ϕs, represents the phase offset between the reference and scattered fields. As the particle size decreases, the pure scattering term decreases rapidly in magnitude such that, for subwavelength nano-objects, the detected irradiance can be approximated as
| 7 |
Unlike darkfield microscopy, the presence of reference light dominates the detected intensity, and any analyte signal appears as a small perturbation in the final image with contrast, c, given by
| 8 |
The potential for ultrasensitive optical detection with interferometric scattering as the contrast mechanism stems from the linear proportionality of the signal to the nano-object volume, as opposed to the square dependence in darkfield imaging. Detection based on interferometric scattering differs from darkfield imaging in two further significant ways. First, the magnitude of the interferometric term is not only determined by the electric field amplitude but also by the phase difference between the interfering fields. Second, the challenge involves the detection of the interferometric term, which is not only much smaller than the reference field but can be lost among unwanted interferometric signals. It is these two parameters that current methods thus focus on: optimizing the phase difference between incident and scattered light and the relative magnitudes of Er and Es, which does not affect the nominal measurement sensitivity but is important for matching the detected light intensity to the capabilities of the detector and the ability to fine-tune the focus of the microscope.
Similar to darkfield imaging and the precursors to modern interferometric detection technologies, the ultimate challenge for ultrasensitive optical detection with interferometric scattering remains the same: eliminating background signatures that overwhelm the signal, in this case the interferometric term. These can be largely attributed to two sources: (1) Additional background light, often due to spurious back-reflections from optics and backscattering from rough detection surfaces. (2) Shot-noise-induced fluctuations in the detected reference light, |Er|2, which can mask a small interferometric signal.
In terms of background scattering, the advantage of interferometric approaches is the weaker, linear, scaling with object volume, meaning that the detected signal drops more slowly for smaller objects. It is important, however, to recognize that this difference in scaling does not make interferometric detection significantly more sensitive than darkfield imaging (within a factor of √2). The main reason why interferometric methods have been more successful than darkfield methods at detecting the smallest objects in recent years comes down to limitations of dynamic range in detection coupled with the unavoidability of a certain level of background scattering as a result of imperfect surfaces. As an example, while it may be feasible to detect a signal that is 1/100 of the background in interferometric imaging, the same challenge in darkfield imaging would amount to detecting a signal that is 1/10000 of the background.
In terms of the signal-to-noise ratio of the measurement, the initial challenge is to ensure that the measurement is mainly limited by shot noise, rather than by other noise sources, such as mechanical vibrations, laser intensity noise, or background fluctuations. Suppressing these additional noise sources becomes a more significant challenge as the shot-noise-induced fluctuations are reduced to the levels required for single-molecule detection. In addition to this, detecting a sufficient number of photons to reach such noise-floor levels might require the signal from detector pixels or recorded frames to be averaged together, thus sacrificing localization precision and temporal resolution, respectively. The temporal averaging of consecutive frames increases the effective full well capacity of the detector, however, as the averaging window is increased, nonshot-noise contributions, such as sample drift, become increasingly prominent. This results in the breakdown of the SNR ∝ √N relationship for larger averaging windows (>50 ms). In this section, we discuss a variety of recent methods that attempt to optimize the phase and amplitude of Er to varying degrees, depending on the specific application and the magnitudes of the noise.
4.2. Interferometric Scattering Microscopy I: Single Particle Tracking
Interferometric scattering microscopy (iSCAT) can, in its simplest form, be thought of as the laser-illumination equivalent of IRM,21,22 in which the stability of a common-path geometry (whereby the interfering light fields share a near-identical optical path to the detector) is coupled with the reference field attenuation and axial sensitivity of a reflection-based configuration. In reflection-based microscopes, the majority of the incident light is transmitted such that the reference field intensity is essentially a measure of the reflectivity at the substrate–sample interface. While this does improve the image contrast compared to that of an analogous transmission-based microscope, it is important to note that it does not improve the overall shot-noise-limited SNR of the experiment, due to the overall photon deficit and the resulting effect on the shot noise. Though the term iSCAT was first coined by the Sandoghdar group,117 its earliest practical implementation was reported using a laser-scanning confocal microscope equipped with a 50:50 beamsplitter imaging microtubules at a glass–water interface.118 The resulting images exhibited a remarkable signal-to-background ratio of ∼10, implying a detection limit on the order of ∼300 tubulin monomers and making this early reflective laser microscope one of the most powerful label-free biosensors of the time. Lindfors et al. then used a reflective laser microscope with sample scanning to demonstrate record detection sensitivity and spectroscopic characterization of GNPs down to 5 nm in oil,119 a concept that quickly expanded to biological environments120 and virus detection and tracking.121 Static detection sensitivity has reached as far as cadmium selenide quantum dots122 and 2 nm gold particles on suitable substrates.123
Two illumination schemes are widely used in iSCAT microscopy: widefield and rapid beam scanning. Widefield illumination uses a lens to focus the illumination into the BFP of the objective, yielding effectively plane-wave illumination of the sample (Figure 7A). This illumination scheme is the method of choice for obtaining the highest possible temporal resolution, as the acquisition rate is not limited by the rate of beam scanning. The second illumination scheme, rapid beam scanning illumination, uses a pair of acousto-optic deflectors (AODs), which scan a focused beam across the sample, generating an even illumination profile while removing ring artifacts and reducing speckle patterns observed in widefield iSCAT (Figure 7B). This illumination approach has often been implemented in combination with a polarization-dependent method to separate the signal from the illumination path using a quarter-wave plate (QWP) and polarizing beamsplitter (PBS), to maximize collection efficiency. The progress of iSCAT over the past decade, as well as the historical context of its development from technological precursors, have been covered in detail in recent reviews.124−127 Here, we will focus on recent technological developments in the context of single-particle tracking (SPT).
Figure 7.
Interferometric scattering microscopy principles and example experimental arrangements. (A) Schematic of widefield illumination iSCAT. (B) Schematic of rapid beam scanning illumination iSCAT. OBJ, objective; BS, beamsplitter (nonpolarizing); QWP, quarter-waveplate; PBS, polarizing beamsplitter; L1, illumination lens; L2, imaging lens; C, camera. (C) Confocal detection iSCAT image of 20 nm GNPs on a glass coverslip in water. (D) Principle of interferometric scattering microscopy, Ei incident field, Es scattered field, Er reflected reference field, Δφ phase difference between the scattered and reflected fields. (C) Adapted with permission from ref (120). Copyright 2006 Optical Society of America.
For SPT, localization precision and imaging speed are ultimately limited by the number of photons that can be detected from the scatterer in a finite time window. Nanoparticles, specifically (GNPs), are commonly used as labels as they are inert, provide a strong extinction signal due to their polarizability, and do not suffer the drawbacks of fluorescent alternatives. Laser-based illumination provides access to incident photon intensities only limited by sample damage and detector full-well depth. As a result, high spatiotemporal resolution can be obtained through a variety of combinations of lateral localization precision and temporal resolution depending on the experimental requirements due to the trade-off between these two parameters. For example, in a recent study, the tracking of 20 nm GNPs was performed with a 2 nm precision, on the μs time scale.128 More recently, this has been pushed toward an acquisition rate of 913 kHz and sub-10 nm localization precision for 20 nm GNPs diffusing on a supported lipid bilayer.129 The sensitivity and measurement precision achievable with iSCAT has enabled optimization of nanoparticle binding130 and the study of mobility differences between supported and suspended bilayer membranes.131
In addition to studying membrane mobilities, iSCAT has recently found applications in elucidating the mechanisms of molecular motor processivity.94,132,133 For example, iSCAT was used to track the position of 30 nm GNP scattering labels attached to the motor domains of kinesin-1 motor proteins, with a localization precision of ∼2 nm at an acquisition rate of 1 kHz.94 More recently, studies have emerged that take advantage of the high spatiotemporal resolution of iSCAT to visualize and characterize the dynamics of cytoskeletal filaments, specifically microtubules.134 Maintaining the imaging capabilities of iSCAT requires highly efficient suppression of background signals, which is relatively straightforward in the aforementioned scenarios but much more challenging in the presence of large objects, such as cell bodies. While in principle, background subtraction approaches can be employed, they are limited to any changes that occur in the background during the desired exposure time, resulting in studies so far being limited to labels of 30 nm diameter or larger.135−137
An additional challenge, and opportunity, of performing iSCAT on cells is the fact that the labels no longer diffuse only in 2D, where the phase relationship between scattered and reflected light is constant, but also in 3D, where it varies rapidly with the distance of the label from the reflective cover glass surface. Early studies used a simple interpretation of eq 6 to extract three-dimensional information both in solution138 and on cells;136 this was recently replaced by a more sophisticated PSF analysis, which makes it possible to achieve sub-10 nm localization precision in both the lateral and axial directions at microsecond acquisition speeds.135 In this way, the authors could study subdiffusion and nanoscale confinement of GNP-labeled epidermal growth factors in live HeLa cells. Additionally, the trajectory of a GNP-labeled protein was used to reconstruct a map of the nanoscopic topology of cellular structures, such as the surface of a filopodium. Subsequent development of interferometric PSF analysis has built upon this work to further demonstrate the applicability of iSCAT to 3D SPT in live, complex environments.137,139 The long observation times of these tracking measurements, up to tens of minutes in length, highlights a key advantage of scattering labels compared to fluorescent alternatives.
Alternative approaches to extend iSCAT detection beyond two-dimensional imaging have been explored by Hong, Cho, and co-workers.140 Remote focusing iSCAT (RF-SCAT), in which imaging depth, and thus the vertical tracking range, is increased by vibration-free vertical image scanning. By implementing an additional 4f telecentric system, the wavefront from the BFP of a high-NA, oil immersion objective, coupled to the sample, can be reimaged into the BFP of a lower-NA, air objective, before being focused onto a reference mirror. The vertical image sections can be selected without movement of the sample stage or imaging objective by translating the axial position of the reference mirror. This optical configuration enabled a trackable depth for nanoparticles of 10 μm.
The ability to use iSCAT in three dimensions with high levels of sensitivity is important because it removes the need for immobilization on a surface, which can be as perturbative, if not more perturbative, than the incorporation of fluorescent labels. In this regard, the combination of the anti-Brownian electrokinetic (ABEL)141,142 trap with iSCAT, termed ISABEL, enabling trapping and closed-loop feedback tracking of nanoscale particles in solution is particularly attractive (Figure 8).143 A challenge compared to other nanoparticle applications arises from the fact that the trapped object is not bound to the glass surface and can move along the optical axis, which causes changes in optical contrast (Figure 7D). The ABEL trap then ensures that the particle is constrained to the trapping region (Figure 8A), through tracking its position with detection rates up to 10 kHz, calculating the particle displacement vector from the center and applying an electric field to move the particle back to the center of the trap (Figure 8B,C). In addition to the ISABEL trap, various other single-particle traps have utilized iSCAT detection as an alternative to fluorescence.138,144−148 The combination of iSCAT detection and single particle traps represents an important advance, especially in the context of the potential future expansion toward single proteins32,33 and combination with motion control and sorting.147,148 The incorporation of iSCAT detection in a nanofluidic rocking Brownian motor capable of sorting GNPs into populations based on their diameters was presented by Knoll and co-workers.148 The device used a combination of deep and narrow ratchets angled to the left, embedded within wide and shallow ratchets angled to the right (Figure 8D). This enabled the separation of 60 and 100 nm GNPs upon application of an external oscillating electric field, with the 60 nm GNPs traveling along the shallow channel to the right and the 100 nm particles traveling along the deep channel to the left (Figure 8E–G). The position of the nanoparticles in the device was imaged using iSCAT at an acquisition rate of 1 kHz.
Figure 8.
Combination of interferometric scattering detection and single particle traps. (A–C) Anti-Brownian electrokinetic trap (ISABEL). (A) Illustration of the scanning pattern of the illumination beam. (B) Schematic of the trapping algorithm. (C) Trace of iSCAT contrast with time of a trapped 40 nm GNP. (D–G) Nanofluidic rocking Brownian motor capable of sorting GNPs based on their size. (D) Topography of the sorting device. (E) Schematic of the method of separation of 60 and 100 nm GNPs. (F) iSCAT images displaying the separation of 60 and 100 nm GNPs. (G) Plot of position vs median relative brightness of the particles in the device before and after sorting. (A–C) Adapted with permission from ref (143). Copyright 2019 American Chemical Society. (D–G) Adapted with permission from ref (148). Copyright 2018 The American Association for the Advancement of Science.
4.3. Interferometric Scattering Microscopy II: Label-free Imaging
The enhanced interferometric signal generated by the plasmon resonance of GNPs is roughly equivalent to that of a dielectric particle with about twice the diameter of a GNP. Given the size range of many biomolecules, for example, virus particles (20–200 nm), iSCAT is well-suited for imaging such objects without the need for labels. Initial studies reported the label-free optical detection of single virus particles bound to supported membrane bilayer receptors.117,121 Manoharan and co-workers also tracked the position and orientation of unlabeled bacteriophages to nanometer precision and monitored the changes in iSCAT contrast as a function of virion DNA content.149 This allowed the DNA ejection dynamics of individual bacteriophages, upon the addition of LamB receptor, to be investigated. Subsequently, the same group quantified the assembly kinetics of individual MS2 bacteriophage capsids around MS2 RNA, identifying features of capsid growth that would otherwise be unresolved in bulk measurements.150 The iSCAT signal intensities of the RNA genome of MS2 bacteriophages, tethered to a functionalized glass surface, were monitored upon the introduction of MS2 coat-protein dimers. The ratio of the iSCAT signal to the number of coat proteins bound to the RNA strand was sufficient to infer potential assembly pathways.
Another target for label-free iSCAT have been microtubules, as in the original study by Amos and Amos.118 In that study, the authors imaged microtubules bound to standard microscope cover glass. This was later extended to gliding assays employed by Andrecka et al.151 As microtubules exhibit anisotropic scattering profiles, their iSCAT contrast is polarization-dependent relative to their orientations, a property previously explored for GNRs in both iSCAT and darkfield modalities.75,77,99,152 Parallelized polarization-sensitive iSCAT detection recently enabled studies of not only microtubule motion but also disassembly.153 Differential image contrasts from the two orthogonal polarization projections (Figure 9A) were presented in the form of a composite kymograph (Figure 9B) to show the change in relative isotropic and anisotropic scattering contributions, indicating a mass change at the microtubule tip (Figure 9C). Incremental images, temporally separated by 19 ms, provided quantitative information on disassembly rates and were used to reconstruct super-resolution images which enabled detection of tubulin oligomer detachment, with a theoretical detection limit of ∼3 tubulin dimers (Figure 9D). Obtaining the differential contrast from the two orthogonal projections (Figure 9A) enabled reconstruction of super-resolved images and nanoscopic structural changes at the tip of a disassembling microtubule from changes to scattering anisotropy on a submillisecond time scale.
Figure 9.
Interferometric scattering microscopy of microtubules and type IV pili. (A) Images of a microtubule acquired with light polarized parallel (left) and perpendicular (right) to the microtubule. Scale bar: 1 μm. (B) Composite kymograph of isotropic (red) and anisotropic (green) scattering combined. Scale bars: 1 μm (vertical) and 1 s (horizontal). (C) The profile of the mean contrast (black) and anisotropic contrast (red) averaged from disassembling microtubules. (D) Cross sections of the iSCAT (top left) and super-resolved (top right) microtubule images inset. (E–H) iSCAT images of different steps in the type IV pili cycle of a Pseudomonas aeruginosa cell, scale bar = 2 μm. (E), (F), (G), and (H) correspond to extension, attachment, tension, and detachment, respectively. (A–D) Adapted with permission from ref (153). Copyright 2021 John Wiley and Sons. (E–H) Adapted with permission from ref (154). Copyright 2019 Springer Nature.
Persat and co-workers transitioned iSCAT imaging of biological filaments toward the cellular context.154 To achieve this, they imaged type IV pili (TFP), filaments assembled at cell surfaces used by bacteria to attach to surfaces, move and resist flow. These filaments have been challenging to image using light microscopes due to the detrimental effects of fluorescent labeling on the cell’s ability to (dis)assemble the filaments, which is central to their function. Using iSCAT, the filaments could be imaged in a label-free fashion (Figure 9E–H), despite the strong scattering background from the cell body. In addition, the dependence of the iSCAT signal on the distance from the glass surface made it possible to differentiate between different functional states, such as extension (Figure 9E), attachment (Figure 9F), development of tension (Figure 9G), and eventually detachment (Figure 9H).
The ability to detect and image biological nanoparticles in aqueous solution naturally extends to lipids and the structures they form such as lipid nanodomains,155 lipid bilayers,156 and micelle and vesicle formation during autocatalytic processes.157−159 iSCAT is particularly well-suited to this application, as it enables label-free visualization of chemical reactions by monitoring the production or destruction of nanoparticles. While nanoparticle formation can be monitored by bulk methods such as dynamic light scattering (DLS)160,161 or NMR, the ability to detect individual particles provides access to the earliest stages of nanoparticle formation, often part of the “lag phase” caused by the invisibility of particles at very low concentrations for bulk methods. In addition, single particle detection allows much improved characterization of sample hetero/homogeneity compared to methods providing average particle size only.
4.4. Mass Photometry
At the very extremes of sensitivity, iSCAT has been used to detect, image, localize, and track individual proteins in a label-free fashion.32,33,162,163 The interferometric contrast generated by protein molecules, which are weakly scattering objects, is a function of their refractive index and proportional to their volume. Given that proteins consist of amino acids, which exhibit small variations in refractive index164 and specific volume,165 the refractive index contribution to the contrast should be effectively constant, and the volume should be proportional to the mass, resulting in a linear relationship between iSCAT contrast and protein mass. The earliest demonstrations of single-protein iSCAT sensitivity used this relationship to provide evidence for single-molecule sensitivity.32,33
Given the enormous power of native mass spectrometry (MS) in the context of studying biomolecular structure and dynamics, label-free single molecule detection, and mass quantification have immense potential.166,167 Early implementations,32,33 however, failed to provide the resolution, accuracy, or precision exceeding that of well-established ensemble-based scattering approaches such as DLS.160,161 Solutions of proteins of different mass could be distinguished, but the resolution was insufficient to quantify mixtures of different species, and the accuracy insufficient to confidently identify species directly by mass, which are some of the key advantages of native MS.
The challenge in nonfluorescent single-molecule imaging relates to the capability of the optical system to distinguish the weak analyte signal from the cacophony of background light contributions. Imaging in a reflective configuration and applying a sliding window background subtraction algorithm to subtract static background can go some way toward addressing this issue.43 However, a typical iSCAT measurement of single proteins in solution yields image contrasts of approximately 0.4%/MDa of molecular mass.32,33 Thus, a perfectly shot-noise-limited measurement of a 250 kDa protein, with an SNR of 10, would require the collection of 108 photoelectrons. Consequently, because of the full-well-capacity limitations of commercially available cameras (<105), this photoelectron requirement must be fulfilled by means of spatiotemporal averaging, limiting resolution and potentially introducing additional background noise.
In the context of biomolecular mass measurements, and the accuracy and mass resolution demanded by applications in bioanalytics, at least an order of magnitude improvement in measurement precision at the single molecule level was required. One method to achieve this involves further attenuation of the reference light relative to the scattering signal by exploiting the geometric separation of the two fields, as described in section 2. The placement of a partially reflective metallic film of specified dimensions in the BFP of the microscope objective selectively filters its NA and increases the scattering contrast (Figure 10A–D).43−45 Again, this does not lead to an improved shot-noise-limited SNR, due to the concomitant reduction in total detected photoelectrons. However, two significant advantages arise from such NA filtering. First, the increased contrast of the substrate surface enables facile and accurate determination of the optimum focus position, which is essential for accurate contrast measurement because of the importance of the phase difference between scattered and reflected light. The second advantage is the ability to use high illumination powers without saturating the imaging detector. However, with the increased contrast provided by the partial reflector, sensitivity to non-Poissonian image perturbations also increases. This is particularly relevant because visualization of single-molecule events require subtraction of the high-contrast, static, speckle-like pattern generated by the surface of a typical glass coverslip (Figure 10D). The increased need for sample stage stability and overall suppression of low-frequency environmental noise resulted in the evolution of a robustly enclosed mass photometer from previous, optical-table-based iSCAT microscopes.34
Figure 10.
Numerical aperture filtered iSCAT. (A) Schematic of the dipole emission pattern for a point scatterer at a refractive index interface; n0 and n1 represent the refractive indices of the media (n1>n0). (B) Emission pattern at the back aperture of a high-NA (1.42) microscope objective for a nanoscopic scatterer at a glass–water interface illuminated with circularly polarized light. The inner outlined circle indicates the position of the partial reflector (top panel). Normalized emission density as a function of numerical aperture. Gray shaded region corresponds to the position of the partial reflector (bottom panel). (C) Raw iSCAT image of a glass coverslip covered by water. (D) Equivalent image with the addition of a T = 1% partial reflector in the optical setup. Scale bar: 1 μm. (E) Illustration of a photothermal spatial light modulator (PT-SLM). (F) XY cross-section of the temperature profile across the PT-SLM (top left panel), equivalent XZ cross-section (top right panel). Scale bar: 100 μm. The two white dashed lines indicate the boundaries of the thermo-optic material. XY cross-section of the resulting refractive index variation (bottom left panel) and equivalent XZ cross-section (bottom right panel). (G) Phase-shift profile of a plane wave propagating through the structure in z at the position of the red dashed line in (I). (H,I) Normalized iSCAT images of 30 nm single gold nanospheres at total heating powers incident on the modulator structure of 0 mW (L) and 110 mW (M). Scale bar: 500 nm. (B–D) Adapted with permission from ref (43). Copyright 2017 American Chemical Society. (E–I) Adapted with permission from ref (168). Copyright 2021 Springer Nature under Creative Commons Attribution 4.0 International License https://creativecommons.org/licenses/by/4.0/.
Recent developments in the Piliarik group expanded the general principle of introducing a spatial filter beyond static field attenuation or phase retardation.168 Replacing the thin metal film disk with a layer of GNRs, the purpose of the spatial filter was not solely to attenuate the reference field but also to change its phase. By sandwiching a layer of thermo-optic material, namely liquid glycerol, between the gold-coated glass substrate and a sapphire heat sink (Figure 10E), the local refractive index of the glycerol can be rapidly altered by microscopic heating (Figure 10F), which in turn results in a phase retardation of the transmitted optical wave (Figure 10G). The refractive index control, spatially confined to the cross-section of the photothermal spatial light modulator (PT-SLM), allows the phase offset between the reference and scattered field to be modulated with a switching time of 70 μs, a factor of 100 faster than conventional liquid crystal SLM devices. This novel device, incorporated into an iSCAT optical setup, was shown to enable quantitative phase imaging measurements of 3D nanoscopic microtubule displacement.
The application of NA filtering in combination with improvements in the hardware and data analysis led to rapid advancements in the detection sensitivity, precision, and ultimately mass resolution of single-molecule mass measurements.34 The contrast distributions from a sample of bovine serum albumin (BSA) molecules (66 kDa) now exhibited well-resolved (19 kDa full width at half-maximum (FWHM)), equally spaced peaks as expected for different oligomeric states of the same protein measured using a method sensitive to complex mass (Figure 11A). In addition, mass measurement of a wide range of single protein molecules was possible with up to 2% sequence mass accuracy. This level of accuracy was a desirable aspect of mass photometry (MP) because it dramatically simplifies the identification of unlabeled species by mass, enabling more highly multiplexed detection than is possible with fluorescence labeling. While the lower detection limit of MP (∼40 kDa)169 is limited by the noise sources discussed previously, the upper limit is dictated by a number of factors. The linear relationship between iSCAT contrast and mass relies on the assumption that the amplitude of the scattered field is negligible when compared to the reference field, which can be somewhat alleviated by altering the degree of reference field attenuation. In addition to this, as the physical size of the particle exceeds the limits of Rayleigh scattering (∼10% of the illumination wavelength) and beyond, both the emission profile and relative phase of its scattered field will begin to deviate from that of a single molecule. However, in the context of single proteins and other similarly sized biomolecules, given an appropriate mass standard, there is no intrinsic upper limit to MP quantification.
Figure 11.
Mass photometry principle and applications. (A) Scattering contrast histogram acquired from binding events of 12 nM BSA. (B) MP images of a BSA monomer, dimer, trimer, and tetramer, scale bars = 200 nm. (C) Contrast to mass relationship for a range of proteins. (D,E) Relationship between contrast and number of base pairs in double-stranded (D) and single-stranded (E) DNA. (F) Mass histogram of Escherichia coli bo3 oxidase isolated from lauryl maltose-neo-pentyl glycol detergent micelles. (G) Mass histograms of different ratios of Escherichia coli outer membrane protein trimers solubilized in amphipols. (A–C) Adapted with permission from ref (34). Copyright 2018 American Association for the Advancement of Science. (D,E) Adapted with permission from ref (175). Copyright 2020 Oxford University Press under Creative Commons Attribution 4.0 International License https://creativecommons.org/licenses/by/4.0/. (F,G) Adapted with permission from ref (184). Copyright 2020 Philipp Kukura.
While it is important to note that the current capabilities of MP cannot match those attainable by native MS, the ability to separate different oligomeric states, as well as free and bound complexes as a result of the high single-molecule measurement precision, in combination with label-free detection, single-molecule sensitivity, mass measurement, solution operation, and concentration measurement by molecular counting has resulted in significant adoption of MP since its inception. This includes application to nucleic acids (Figure 11D,E), membrane proteins (Figure 11F,G), and membrane associated proteins.34,158,170−197 The study of membrane proteins in particular presents an experimental challenge due to the instability of their hydrophobic transmembrane regions in aqueous solution. In the first study applying MP to study membrane proteins, the authors studied both amphipathic vehicles and lipid nanodiscs in order to characterize the particle size, sample purity, and heterogeneity of integral membrane proteins, in addition to identifying their functional states.184 MP was applied to imaging different oligomeric states of Escherichia coli bo3 oxidase isolated from lauryl maltose-neo-pentyl glycol detergent micelles (Figure 11G). In addition, the authors studied different ratios of Escherichia coli outer membrane protein (OmpF) trimers solubilized in amphipols (Apols) (Figure 11F).
The speed, sensitivity, and relative simplicity of a typical experiment mean that MP lends itself naturally to sample characterization for more structure-specific analytical techniques such as cryo-electron microscopy (cryo-EM) and X-ray crystallography.193 A recent study demonstrated this by using MP to quantify the heterogeneity of macromolecular complexes. The results obtained were comparable to those gathered from corresponding negative stain EM workflows. Similarly, the ability of MP to count individual molecules can be used to accurately determine the relative abundances of different biomolecular complexes in solution, thus yielding information on the associated binding affinities and interaction kinetics.187,194,195
4.5. Interferometric Reflectance Imaging Sensor
Interferometric reflectance imaging sensor (IRIS) is an experimental technique combining interferometric imaging principles with a specialized substrate to act as an effective biosensor. In IRIS, the substrate is fabricated from a silicon wafer covered by a thermally grown layer of silicon dioxide, which can be coated with antibodies to facilitate selective binding of a species of interest.198 Initially, the technique was designed to detect ensemble binding of biomolecules to a surface by measuring the thickness of the accumulated layer of biomass.199 Subsequently, the technique demonstrated the ability to detect and image single particles, in a modality termed single particle interferometric reflectance imaging sensor (SP-IRIS),200 which is the focus of this section.
In SP-IRIS, the SiO2/Si substrate is imaged using a widefield reflection microscope containing a beamsplitter and a light-emitting diode (LED) illumination source (Figure 12A).200 LED illumination provides a simple method to suppress coherent image artifacts, such as speckles. The reference field is generated from the light reflected by the IRIS substrate, hence the phase difference between the scattered and reflected field is optimized by tailoring the thickness of the SiO2 layer.201 In addition, the IRIS substrate increases the collection efficiency of the scattered light from a nanoparticle when compared to that of a transparent substrate by reflecting some of the forward scattered light from the particle. In the original working mode of SP-IRIS developed by Ünlü and co-workers, nanoparticles were immobilized on the substrate surface before being imaged with a high-NA air objective lens. Proof of principle experiments demonstrated the ability of SP-IRIS to detect and characterize the size of polystyrene beads immobilized on the substrate surface, ranging from 70 to 200 nm in diameter, and H1N1 influenza virus particles ranging from 90 to 160 nm in diameter (Figure 12B,C).
Figure 12.
Single-particle interferometric reflectance imaging sensor (SP-IRIS). (A) Schematic illustrating the experimental configuration: OBJ, objective; BS, beamsplitter. (B) Image of H1N1 virus particles immobilized on an SP-IRIS chip. (C) Histogram of the measured H1N1 virus diameters. (D) Composite image of an SP-IRIS microarray. Inset: normalized image intensity image of 25 nm × 71 nm GNRs bound to the surface of a microarray spot. (E) Plot of GNR counts against the concentration of HBsAg. (B,C) Adapted with permission from ref (200). Copyright 2010 American Chemical Society. (D,E) Adapted with permission from ref (202). Copyright 2018 American Chemical Society.
Building on this initial work, the application of SP-IRIS was extended to multiplexed detection of different viruses in complex media.203 The IRIS chip was coated with a nonfouling copolymer functionalized with an array of multiple different monoclonal antibodies, which selectively bind to different virus surface glycoproteins. In this study, the SP-IRIS chips were incubated in the sample and were washed and dried before being imaged. The authors demonstrated that the technique was capable of detecting virus particles in both blood and fetal bovine serum (FBS). In addition, SP-IRIS was able to differentiate between multiple vesicular stomatitis virus (VSV) based virions in a mixed sample due to the size variations between different species. This capability was developed by Connor and co-workers, who used two LED illumination wavelengths to detect virus particles over a range of 40 to ∼400 nm in diameter.204 A similar approach was used to detect exosomes by functionalizing the surface of the IRIS chip with a microarray of antibodies which selectively bind to exosomal biomarkers.205 The technique was applied to detect exosomes with diameters >50 nm from purified samples and undiluted human cerebrospinal fluid.
SP-IRIS transitioned from operating in air to operating in buffer, enabling real-time measurements.206 To overcome the reduced signal when operating in buffer, image processing techniques and a higher-NA air objective lens were introduced, along with optimization of the SiO2 thickness for these conditions. In this implementation, SP-IRIS demonstrated real-time detection of an Ebola model virus binding to an antibody-microarray coated IRIS chip in FBS with a detection limit of 100 PFU/mL. In the experiments presented, the temporal resolution of the technique was 30 s, which was adequate due to the low binding frequency of the viruses at the concentrations of interest. For measurement at higher concentrations, the temporal resolution could be improved through the use of a faster camera. This concept was extended by Scherr et al., who developed a disposable cartridge suitable for use with infectious samples, enabling SP-IRIS to act as a potential point-of-need diagnostic technique.207 Moreover, the capture efficiency of virus particles in SP-IRIS was improved by incorporating advanced surface functionalization techniques and assay development, yielding a limit of detection down to 43 PFU/mL.208,209
SP-IRIS has undergone multiple recent technological developments. First, a number of strategies capable of analyzing stacks of defocused SP-IRIS images were developed, which improves the visibility of nanoparticles in SP-IRIS images.210−212 Moreover, SP-IRIS adopted a similar approach to differential phase contrast213 and Fourier ptychographic microscopy81,214 by combining asymmetric illumination with computational reconstruction to improve the spatial resolution of the acquired images.215 The technique was applied to imaging an Ebola model virus immobilized on an SP-IRIS chip surface in air with a spatial resolution of ∼150 nm, demonstrating a factor of 2 improvement over standard SP-IRIS.
In addition to the label-free imaging applications described above, SP-IRIS measurements have also included scattering labels. In a demonstration of this concept, β-lactoglobulin proteins were detected by combining SP-IRIS with antibody-functionalized 40 nm GNPs.216 Antibody-functionalized SP-IRIS chips were incubated with media spiked with β-lactoglobulin, enabling the protein to bind to the chip. Following this step, the chip was incubated with antibody-functionalized 40 nm GNPs, enabling the GNP labeled protein complex to be detected using SP-IRIS. The calculated limit of detection of the technique was ∼60 aM in undiluted serum and ∼500 aM in unprocessed blood. Furthermore, SP-IRIS used GNR labeling to detect and count viruses over a large FOV.202 As GNRs scatter light which is linearly polarized along their longitudinal axis,37,76 the authors used circularly polarized illumination in combination with polarization optics in the detection to selectively attenuate the reflected field relative to the scattered field. This enabled the detection of 25 nm × 71 nm GNRs while using a low-NA, low-magnification objective lens with a FOV of 1.46 mm2, which enabled tens of GNR-containing microarray spots to be imaged simultaneously (Figure 12D). Because of the relationship between the orientation of the GNR and the retardation of the scattered field imposed by the polarization optics in the detection, a z stack of images was acquired and processed so that GNRs of all orientation angles were detected. This modality of SP-IRIS was used to detect hepatitis B surface antigen (HBsAg) in buffer, using 15 nm × 40 nm GNRs as scattering labels. Similar to the study outlined above, an antibody-microarray coated SP-IRIS chip was incubated with HBsAg, followed by incubation with antibody-functionalized GNRs. After these steps, the SP-IRIS chip was imaged in air to quantify the amount of GNR-labeled HBsAg bound to the antibody-microarray coated IRIS chip (Figure 12E). Combining single particle counting and inferring the number of nanorods within an unresolvable distance by analyzing the intensity of the signal, the sensor was capable of operating over a dynamic range of ∼1 × 106, with a limit of detection of 3.2. pg/mL. This work was extended to dynamic measurements by using a custom perfusion chamber and a higher-NA objective, sacrificing the size of the FOV in exchange for higher collection efficiency of scattered light.217 In addition, an analysis scheme which tracked each binding event over time was developed so that the rate of binding and unbinding in a sample could be determined.
4.6. Coherent Brightfield Imaging
In coherent brightfield imaging (COBRI), the interference takes place between forward scattered light from the object of interest and the transmitted illumination light. COBRI was introduced by Hsieh and co-workers, who showcased the method by label-free imaging and tracking of vaccinia virus particles (∼250 nm in diameter) diffusing on a cell plasma membrane, at a frame rate of up to 100 kHz and a spatial precision of <3 nm (Figure 13B–E).218 The authors demonstrated that the method is capable of three-dimensional tracking as the axial position of the object can be retrieved by analyzing the PSF due to the variation in the phase difference between scattered and transmitted light fields with the axial position of the scatterer. COBRI exhibits a single contrast inversion as the object of interest is translated through the focus, making axial tracking particularly intuitive and straightforward (Figure 13C). To track virus particles with high localization precision in the presence of scattering background from the cell membrane, the authors employed a digital background removal procedure. The experimental setup used a pair of AODs to scan a spatially filtered laser beam across the sample to generate an even illumination profile. The original experimental design of COBRI incorporated a 60 mm focal length lens to focus the illumination onto the sample, although this was later replaced by a water-dipping objective lens (Figure 13A).219
Figure 13.
Coherent brightfield microscopy. (A) Schematic illustrating the experimental configuration. OBJ1, water-dipping objective; OBJ2, oil-immersion objective. (B) Image of vaccinia virus particles immobilized on a glass coverslip. (C) Image of a vaccinia virus particle as a function of axial sample position. (D) 3D localization of a tracked vaccinia virus particle immobilized on a glass coverslip at a frame rate of 5 kHz. (E) 3D trace of a virus particle landing and diffusing on a cell plasma membrane at an acquisition rate of 5 kHz. (B–E) Adapted with permission from ref (218). Copyright 2017 American Chemical Society.
COBRI microscopy was further used to study intracellular cargo transport by imaging and tracking cellular vesicles in three dimensions inside live fibroblast cells at a frame rate of 30 kHz.220 Because of the sensitivity limit of the particular setup used, vesicles below 150 nm in diameter could not be observed and hence the study focused on larger vesicles, with a mean diameter of ∼320 nm. Because of the high temporal resolution and long observation times enabled by the technique, transient bidirectional motion of the vesicles inside the cells were observed. In addition, correlated motion was observed by simultaneously tracking two adjacent vesicles. COBRI is particularly well-suited for tracking objects within cells, as the technique does not detect reflections from interfaces within the sample, such as a cell membrane.
Recently, the sensitivity of COBRI was improved so that GNPs of diameters down to 10 nm could be imaged and tracked in water.219 The gain in sensitivity was obtained by employing the reference field attenuation approach discussed in section 4.4.43−45 As a proof of principle, a lipid labeled with a 10 nm GNP was tracked while diffusing on a supported lipid bilayer with an acquisition rate of 1 kHz and localization precision of 10 nm. In addition, the authors built a combined iSCAT-COBRI microscope to enable a quantitative comparison between the two techniques using the same detection line, image processing, and experimental parameters. To obtain a comparable level of camera counts with the same illumination intensity, the transmissivity of the partial reflector was tailored for each technique. It was determined that the sensitivity of the techniques are identical for equal illumination intensity, as expected from eq 7.
5. Plasmonic Imaging
5.1. Introduction
Many bioanalytical technologies leverage surface plasmon resonance (SPR) to amplify the evanescent electric field at the sample–substrate interface, thus enhancing the detected signal emanating from an analyte molecule in the far-field. SPR spectroscopy has been used in biosensing since 1983,221 with the first biosensor based on the optical excitation of surface plasmons demonstrated in the early 1990s by Lundstrom et al. Modern SPR biosensors enable highly sensitive, often label-free investigation of the interaction between biomolecules, such as proteins222−229 and nucleic acids,230−238 with multiple reviews having documented the expansion of the field in great detail.239−245 The majority of these approaches are ensemble-based, integrating the signal from multiple interactions in a macroscopic area so they fail to resolve the range of stochastic behavior present in a population of biomolecules. Recent developments have thus attempted to combine the information available from ensemble-based spectroscopic detection with high-resolution optical microscopy in an attempt to reach single-molecule sensitivity by SPR microscopy (SPRM).
Surface plasmon resonance refers to the collective oscillation of free conduction electrons near the interface of two media with dielectric constants of opposite signs, for example, a metal and a dielectric. The associated electromagnetic field is localized to this interface and decays into both media. SPR spectroscopy measures changes in the optical reflectivity of the interface between these two media, which result from changes in the local refractive index caused by target species entering or leaving the evanescent field. The sensitivity of the measurement to small changes in the local refractive index is enhanced by the excitation of surface plasmon polaritons (SPP); charge density waves formed at, and propagating along, the interface between a metal and a dielectric medium. Excitation of SPPs on a continuous metallic film (typically silver or gold) with incident light requires matching the respective wave vectors. This can be achieved by tuning the angle of incidence of illumination. Consequently, as the illumination angle approaches the resonance angle, excitation of SPPs results in a sudden and sharp attenuation of the reflected light intensity. The resonance angle that leads to this intensity dip is therefore highly sensitive to the effective refractive index (neff), the weighted average of the index of refraction of any materials within the evanescent field (∼300 nm from the metal surface), such as liquids and absorbates.246
A typical SPR instrument excites surface plasmon modes on unstructured metal films by attenuated total reflection (ATR), passing a p-polarized monochromatic light beam through a prism, which is optically attached to a gold-coated glass slide. The reflected light intensity can then be quantified as a function of wavelength, at a fixed angle of illumination, or versus illumination angle for a range of wavelengths. The first demonstrations of spatially resolved plasmonic measurements were published in the 1980s by Yeatman and Ash247 and Rothenhausler and Knoll.248 Initial developments simply replaced the detector in an SPR spectroscopy setup, typically a photodiode, with a digital camera to obtain a two-dimensional image of the local refractive index change on the surface of a thin gold film. This technique is commonly known as SPR imaging (SPRI) or two-dimensional SPR (2D-SPR). Prism-based SPRI platforms provide sufficient spatial resolution to quantify the optical response of multiple cells independently249 and, as a result, they provide a useful tool for several high-throughput and multiplexed analyses in biological research and drug discovery applications.250−254 The spatial resolution achievable when a prism is used to collect the detected light, however, is limited. Replacing the prism with a high-NA objective lens greatly enhances the achievable spatial resolution to that of a wide-field microscope, thus facilitating SPRM.255
In this section, we will focus on recent developments in objective-based plasmonic imaging, particularly those which leverage SPR on continuous metallic films to enhance the signal and elevate plasmonic imaging toward single-molecule detection sensitivity.
5.2. Surface Plasmon Resonance Microscopy
A typical SPRM optical system focuses a beam of p-polarized monochromatic light into the BFP of a high-NA oil-immersion objective, producing plane-wave illumination of a gold-coated glass coverslip, again based on the ATR method, akin to the Kretschmann configuration.256 Lateral offset of the focused beam away from the optical axis of the BFP, d, alters the incident illumination angle, θ, while radial translation changes the azimuthal illumination angle and therefore the direction of SPP propagation. Given the focal length of the objective lens, f, the incident angle of illumination can be determined from eq 9:
| 9 |
Because of imperfect alignment and aberrations in the objective, however, a Fresnel-based optical model may not produce accurate results, thus a calibration measurement may be required. Modulation of the incident illumination angle determines the proportion of light reflected off of the gold–medium interface and collected by the objective before being imaged onto a CCD camera with a tube lens (Figure 14A). The reflected light intensity is strongly attenuated upon excitation of the surface plasmon. Wave vector matching of the incident light to that of the surface plasmon can be achieved by illuminating the metal/dielectric interface at the resonance angle or SPR angle, θR, given by257
| 10 |
where ε1 and ε2 are the dielectric constants of the dielectric medium (typically a buffer solution) and the objective lens, respectively, and εm is the real part of the dielectric constant of the metal film. This equation is valid if the immersion medium and objective lens refractive index are matched. Small refractive index variations in the evanescent field at the interface alter the resonance angle. A practical implication of eqs 9 and 10 is the challenge associated with illuminating a sample at the resonance angle. For example, θR = 71.6° for a 50 nm thick gold film illuminated with λ = 638 nm light at a water interface255 and, as a result, objectives with NA > 1.4 are required. In addition, as θR for gold and silver films typically increases as λ decreases in the visible spectrum, illumination is generally restricted to red light in SPRM experiments, as higher index materials would be required to achieve θi = θR at shorter wavelengths.
Figure 14.
Surface plasmon resonance microscopy. (A) Schematic of a typical SPRM optical setup. OBJ, objective; BS, beamsplitter; L, tube lens; C, camera. (B) An example of a SPRM PSF with the arrow indicating the direction of surface plasmon propagation. (C) Near-field light–matter interaction of SPRM illumination. Ei incident field, Es scattered field, Er reflected field, Esp evanescent field, θR resonant angle. (B) Adapted with permission from ref (258). Copyright 2018 American Chemical Society.
For a uniform surface, measuring the change in reflected light intensity at a fixed incident angle of illumination enables the change in the effective refractive index of the interface to be determined. This is the basic principle of SPR spectroscopy. Resolving discrete local refractive index differences caused by the presence of a nano-object near the surface, however, requires a deviation from the classical model. This heterogeneity in the complex dielectric generates the contrast of an SPRM image. The incident p-polarized light at the substrate/metal interface is partially reflected and absorbed, exciting the surface plasmon wave and resulting in two fields: the reflected field, Er, and the field associated with the surface plasmon, Esp. The propagating surface plasmon wave is scattered by any dielectric discontinuity, such as an object near the metal surface, producing a scattered field, Es (Figure 14C). The light intensity detected at any given point in an SPR image, ISPR, is given by a superposition of the reflected, scattered, and surface plasmon fields, radiated in the direction of reflection:259
| 11 |
The first term in eq 11 describes a uniform SPR background in the absence of a particle. The second term in the expression is a pure scattering contribution, which is negligible for particles much smaller than the illumination wavelength. The complex third term describes the interferometric contribution between the scattering field and the surface plasmon, which can be expressed as an exponentially decaying cylindrical wave and gives rise to a parabolic “tail” pattern of fringes at the image plane (Figure 14B).260−262
Excitation of surface plasmon modes propagating across a continuous metallic surface produces an electric field exhibiting a characteristic propagation length (∼101 μm) and penetration depth (∼102 nm). While this is the most commonly excited surface plasmon mode in SPRM experiments, these properties can be manipulated using conductive-dielectric multilayers,263 exciting localized surface plasmons (LSP) supported by a single nanoparticle,264−267 or by manufacturing arrays of plasmonic nanoparticles268,269 or nanoapertures270−272 on a metallic film. There have been many optical platform developments in the field of spectral SPR imaging and biosensing, with nonresonant detection sensitivities reaching the single molecule level in two independent studies in 2012. Zijlstra et al. employed a sensitive photothermal assay to monitor the SPR of a gold nanorod upon binding of single proteins, producing results which exhibited a 700-fold increase in sensitivity over alternative, state-of-the-art plasmon biosensors at the time.266 In a different approach, Ament et al. illuminated a gold nanorod with a white light laser in TIR and employed a darkfield spectroscopy detection modality; this allowed them to resolve the discrete steps in the SPR wavelength caused by single protein binding events on the millisecond time scale.267 These pioneering developments, as well as many other studies which leverage plasmon sensor technologies, have been covered in other reviews.240,273−275 For this review, we will focus on recent developments in objective-based SPRM setups based on the excitation of propagating SPs on continuous metallic films.
The PSF of an SPRM microscope is distorted by the propagating SPP, and thus its orientation is ultimately dictated by the incident (polar) angle of illumination (Figure 14B). Spatial resolution is diffraction limited in the direction transverse to SPP propagation, while extending over the propagation length of the SPP in the longitudinal direction, typically several micrometers. Tao and co-workers were the first to observe this pattern,276 and subsequently proposed a theoretical model to describe the interaction between a propagating SPP and an isolated nano-object.259 Several theoretical models have also attempted to describe this interaction and its role in the formation of a plasmonic image.277 Quantitative analysis of the wave-like PSF, however, has been challenging due to its geometric complexity despite these complex interferometric scattering patterns containing information regarding the properties of the associated nanoparticle. The summed signal intensity over a region of interest (ROI) selected from a PSF can be used to identify both the size of a nanoparticle and its distance from the metal film. The refractive index, and therefore the nanoparticle material, however, cannot be accurately determined by this method. Quantitative interpretations in the frequency domain have further developed the general understanding and analysis of the SPRM PSF.258 The interferometric pattern partly consists of light scattered from the imaged nanoparticle, hence it must depend on parameters such as the refractive index as well as the size of the nanoparticle. Quantitative analysis of this pattern was achieved, both numerically and experimentally, by implementing a parameter, ψ, which describes the phase of the parabolic patterns leading to the elucidation of the relationship between the refractive index of the nanoparticle and its associated pattern.278
The detection of scattered light of a propagating evanescent wave limits the spatial resolution of SPRM. However, the confinement of the surface plasmon to the metal/dielectric interface provides SPRM with extremely high axial sensitivity. The rapidly decaying evanescent field results in an exponential relationship between SPR signal intensity and the axial distance between a particle and the interface. If the decay constant of the SPRM system is known, this relationship can be quantified with a precision of ∼5 nm in the z-direction.279
5.3. Biomolecular Imaging and Detection
Despite its anisotropic PSF, SPRM has been employed to directly image and study both synthetic nanoparticles and biomolecules. The excitation of a propagating SPP amplifies the local electric field and is scattered by any dielectric discontinuity in its path while also attenuating the amount of light reflected from the surface. Both of these effects contribute toward enhanced interferometric contrast in the far field. Furthermore, the confinement of the evanescent field to within approximately 200–300 nm of the gold–medium interface rejects background scattering from out-of-focus particles, much like in the approaches discussed in section 3. Tao and co-workers leveraged this effect to image single H1N1 influenza A virus particles and monitor their interaction on different surfaces (Figure 15A).276 The multiplexing capabilities of SPRM enabled high-throughput analysis of several individual virus particles. By fitting the SPRM signal from differently sized silica nanoparticles (Figure 15B), a proportional relationship was verified between the volume of the particles and the interferometric signal (Figure 15C). By way of this calibration process, and by approximating the refractive index of viral particles as ∼1.48 due to their protein–lipid composition, the volume and diameter of influenza A was found to be 6.8 ± 3.0 × 10–4 μm3 and 109 ± 13 nm, respectively, in good agreement with literature values.
Figure 15.
SPRM detection of single virus particles. (A) SPRM images of H1N1 influenza A virus and three different-sized silica nanoparticles in PBS buffer. (B) Histograms of SPR intensities of silica nanoparticles of different diameters and of influenza A viral particles. (C) Calibration curve of SPR intensity vs particle volume. (A–C) Adapted with permission from ref (276). Copyright 2010 National Academy of Sciences.
The imaging capability enabled by a high-NA objective provides SPRM with single-molecule sensitivity, first demonstrated by Yu et al. in 2014.280 By implementing differential imaging coupled with lateral translation of the image and a deconvolution algorithm, single DNA molecules were imaged. These images were used to measure the length of individual stretched λ-DNA molecules to determine an average length of ∼14.6 μm, in good agreement with fluorescence images and theoretical models. Applications of SPRM to study larger biological structures, such as single-bacterium motion classification281 and visualization of cellular activity and morphology,282−284 further demonstrate the versatility of the technique, but such applications are mostly beyond the scope of this review.285 It is worth noting that the extension of the electric field penetration depth toward cellular length-scales (∼1 μm) can be achieved by depositing a buffer layer with a refractive index similar to that of the sample between the substrate and the metal film.263 This technique, called long-range surface plasmon resonance (LRSPR), has recently been incorporated into an SPRM setup to study cellular micromotion.284
The sensitivity of a SPRM measurement is determined by its ability to detect small variations in the refractive index, which are proportional to the volume of the analyte molecule. Consequently, quantitative detection of small-molecule binding events is a challenge for a conventional SPRM system. In an alternative approach, plasmonic-based electrochemical impedance microscopy (P-EIM) employs an SPRM optical setup to image the electrochemical impedance distribution across the surface while simultaneously measuring the SPR response.286 Instead of measuring changes in current directly, P-EIM uses the gold surface as an electrode and converts local surface charge variations, which result from the modulation of an applied external field, into an optical signal. In comparison to SPRM, P-EIM yields improved image contrast and greater sensitivity when detecting small molecules. P-EIM measures interfacial impedance rather than refractive index changes, therefore the measured signal does not scale with the mass of the analyte molecule. The superior sensitivity, coupled with submicrometer spatial resolution and submillisecond temporal resolution, makes P-EIM a powerful approach for small-molecule detection and quantitative investigation of protein binding kinetics, as well as a variety of other applications.287−289
The labeling of biomolecules with GNPs can also enhance the detected optical response, as has been shown in multiple SPR biosensor studies. Used in conjunction with SPRM, Halpern et al. demonstrated real-time detection of hybridization adsorption of single DNA-functionalized GNPs.290 A novel method to improve the detection capabilities of SPRM toward small-molecule detection combines the axial sensitivity of SPRM with the signal amplification provided by GNPs and the detection sensitivity of P-EIM. Here, GNPs are bound to a gold electrode with a molecular tether and an oscillating electric field is applied normal to the electrode surface, causing the GNPs to oscillate in the z direction. The plasmonic signal from the nano-oscillators is extremely sensitive to the amplitude of their oscillation, such that the z position of each nano-oscillator can be determined with a precision of ∼0.1 nm.291 From the measured amplitude and phase of the oscillation, the charge of each nano-oscillator can be determined with a detection limit of ∼0.18 electron charges in aqueous solution. This sensitivity to nano-oscillator charge, which is altered by the binding of small molecules, was further employed to monitor phosphorylation kinetics.292 An adaptation of this approach used light to drive the nano-oscillators rather than an external electrical current.293 Mechanoresponsive polymers self-assemble on the gold surface and act as both a tether and a motor for the GNPs, as they extend and collapse under periodic photothermal generation. To monitor single-strand microRNA (miRNA), the complementary strand is immobilized in the gap between the GNP and the gold surface, and as the two hybridize to form a rigid double-stranded structure, the oscillation amplitude of the GNP is damped. Using the time trace of the damping signal, real-time monitoring of single-molecule miRNA hybridization can be achieved.
Unlike traditional localized SPR (LSPR) methods, the implementation of nano-oscillators as signal-enhancing devices is not limited to metallic nanoparticles. A recent study has applied the general principles of nano-oscillator SPRM but replaced the GNP antenna with single proteins and the plasmonic surface with a layer of indium tin oxide (ITO) (Figure 16A).294 An alternating electric field is applied to the ITO surface, driving the oscillation of the tethered protein molecules. With an SPRM optical setup, the surface is illuminated at an oblique angle which produces an evanescent wave, the incident light is scattered by the protein and collected by a high-NA objective, and an image sequence is recorded on a CMOS camera. A fast Fourier transform (FFT) is performed on the image sequence to isolate the oscillation frequency of the applied field and extract the oscillation amplitude while rejecting background noise (Figure 16B). The resulting signal has been used to estimate the size (Figure 16C), charge, mobility, and conformation of the tethered protein. This data was then used to produce a two-dimensional plot of mobility vs size for various single proteins and protein–ligand complexes (Figure 16D).
Figure 16.
SPRM imaging of protein size, charge, and mobility. (A) Schematic showing a single protein tethered to an ITO surface by a 63 nm long polyethylene glycol (PEG) linker. (B) FFT image contrast as a function of potential amplitude and a schematic displaying this relation to PEG tether extension; scale bar = 3 μm. The blue dashed line indicates linear and plateau regimes of tether extension. (C) Size determination of protein–PEG complexes (DH) as a function of FFT image contrast change (ΔC). (D) 2D plot of mobility (μ) vs size (DH) of single proteins and protein–ligand complexes. (A–D) Adapted with permission from ref (294). Copyright 2020 Springer Nature under Creative Commons Attribution 4.0 International License https://creativecommons.org/licenses/by/4.0.
5.4. Resolution-enhanced Surface Plasmon Resonance Microscopy
Although imaging of single molecules is possible with conventional SPRM, the complex, wave-like PSF significantly restricts spatial resolution and sensitivity. Attempts have been made to reconstruct a pseudospherical scattering wavefront and thus a symmetric PSF, both optically and computationally.
One approach to improve the spatial resolution is to use light with a shorter wavelength (e.g., λ = 532 nm for Au) to excite the surface plasmons, although this sacrifices sensitivity as the SPR is less well-defined.255 Lateral raster scanning of a focused probe beam across the surface, in a technique known as scanning localized surface plasmon microscopy (SLSPM), has also been shown to achieve improvements to the spatial resolution but at the expense of temporal resolution.295−297 The wave-like appearance of the SPRM PSF is not entirely unique to SPRM. As discussed in section 3.3, illumination of a nano-object by a plane wave incident at an oblique polar angle produces a similarly anisotropic PSF, with tail-like fringe patterns, as the spherical scattered wave and reflected plane wave have a spatially inhomogeneous phase offset.
Using conventional brightfield microscopy, as discussed in section 3.3, Rohrbach and co-workers overcame these fringe patterns by incoherently averaging images recorded with many different azimuthal angles of illumination.85,91 A similar azimuthal rotation illumination (ARI) approach was implemented in an SPRM microscope (SPRM-ARI) (Figure 17A),298 building upon previous attempts to optically improve the lateral resolution in SPRM.299−301 The characteristic tails produced from SP scattering of a nanoparticle on a silver film were effectively homogenized upon image averaging, resulting in a bright, doughnut-shaped PSF (Figure 17B). This shape can be explained by the polarization-matched coupled emission at all incident azimuthal angles of illumination, analogous to the PSF for surface plasmon-coupled emission microscopy (SPCEM).302−304 A polarizer was inserted in front of the camera, such that this doughnut-shaped spot was separated into two bright spots, which can be regarded as the PSF of SPRM-ARI. The benefits of improved spatial resolution and smoothing of interferometric fringe patterns was further illustrated by imaging the complex shape of a winding nanowire (Figure 17C). SPRM-ARI is said to enhance the longitudinal spatial resolution of SPRM by an order of magnitude, and the effective temporal resolution is only limited by the illumination rotation speed (1 ms), as opposed to the lateral movement time limitation in differential SPR. Alternative nonmetallic films for signal enhancement have also been suggested and compared. Using bespoke dielectric multilayers to produce Bloch surface waves (BSW) instead of surface plasmons, illumination wavelength, resonant angle, and polarization state can be tailored (Figure 17D). While SPRM is restricted to illumination with P-polarized light, commonly in the spectral region of 635 nm for Au films due to resonant angle collection, BSW microscopy (BSWM) can operate with S-polarized light of shorter wavelengths (e.g., 532 nm) and with tunable penetration depths (Figure 17E).
Figure 17.
SPRM-ARI imaging and Bloch surface wave microscopy. (A) Schematic illustrating the superposition of the planar illumination wave, incident at differing azimuthal angles, and a circular scattered wave from a nanosphere. The bottom image shows the cumulative propagation direction of the planar wave for azimuthal illumination angles 0–360°, similar to SPRM-ARI. (B) SPRM image of a 50 nm nanosphere on a Ag film (left) and a corresponding image acquired with SPRM-ARI (right). (C) Images acquired analogously to those in B but of a winding polymer nanowire. (D) Schematic displaying the dielectric multilayer substrate used for BSWM. (E) Angle-dependent excitation spectrum where ne is the refractive index of the environment. (A–E) Adapted with permission from ref (298). Copyright 2019 American Association for the Advancement of Science under Creative Commons Attribution-NonCommercial 4.0 International license https://creativecommons.org/licenses/by-nc/4.0/.
A recent study by the Tao group has modified the traditional SPRM method to image the scattering of SPPs in a transmission modality, which they refer to as plasmonic scattering microscopy (PSM).305 In contrast to reflection-based SPRM, the reflected light is not collected, and plasmonic images are produced by the interferometric interaction of light scattered from both the particle and from topographical inhomogeneities of the gold surface (Figure 18A). This allows for higher illumination intensities (∼30 000×), provides a high-contrast image and circumvents the complexity and limited resolution of the SPRM PSF. Instead, the pixel intensities are integrated across the complex Airy disk (Figure 18B). The enhanced sensitivity of PSM was demonstrated by label-free imaging of single proteins. Human immunoglobulin M (IgM) and human immunoglobulin A (IgA) were detected upon binding to the surface (Figure 18B), and their respective sizes were determined (Figure 18C). This identification was further confirmed by distinguishing both proteins via the specific binding of IgA to an anti-IgA coated surface. The latter experiment enabled the quantification of single-protein binding kinetics by digital counting of individual binding events of IgA molecules to an anti-IgA coated surface (Figure 18D,E). Surface coverage results, obtained by multiplying the binding counts detected by PSM by the molecular mass of IgA (385 kDa), were consistent with ensemble SPR measurements, confirming that PSM can be used to measure protein mass. The PSM optical setup was recently modified to include a Kretschmann prism-coupled SPR illumination configuration, while retaining single protein sensitivity and high spatial resolution enabled by transmission detection.306
Figure 18.
Quantitative detection of single molecules by plasmonic scattering microscopy. (A) Schematic illustrating the detection method employed in PSM, in which plasmonic waves scattered by a particle (Es) and any inhomogeneities on the gold surface (Eb) are gathered from the top and interfere to produce an image at the detector. (B) Background and drift corrected PSM image of immunoglobulin M (IgM) molecules. Scale bar: 5 μm. (C) PSM image intensity vs particle diameter. (D) Histograms displaying intensity changes associated with binding and unbinding of individual IgA molecules. (E) Example of the binding behavior of a single IgA molecule. (B–E) Adapted with permission from ref (305). Copyright 2020 Springer Nature.
Computational approaches to reconstruct an isotropic SPRM PSF, without additional temporal limitations or further complication of the optical setup, have also been explored.307 The image reconstruction algorithm proposed applies filters in Fourier space to obtain the scattered field from the measured image, before reconstructing the object image via deconvolution of the scattered field with the SPRM PSF, modeled by a decaying cylindrical wave. This process was demonstrated on 100 nm polystyrene nanoparticles and achieved near-diffraction-limited spatial resolution in both transverse and longitudinal directions of ∼310 nm. However, in the presence of multiple non-negligible scattering events, a more sophisticated algorithm may need to be applied. In another study, this digital image-processing algorithm was employed in an optimized SPRM setup termed interferometric plasmonic microscopy (iPM).308 Diffraction-limited spatial resolution (∼260 nm) enabled the real-time imaging of single exosomes adsorbing to a modified Au surface, which was then used to determine their respective diameters (30–150 nm). This level of sensitivity resulted quantitative measurements of the membrane fusion activity between exosomes and liposomes, as well as the study of the dynamic interaction between exosomes and antibodies.
6. Summary and Outlook
In this review, we have given an overview of recent developments in darkfield, interferometric scattering, and plasmonic microscopy in the context of nanoparticle and molecular imaging, detection, and characterization. These three approaches have achieved significant increases in sensitivity, with the latter two reaching the single (bio)molecule level under ambient conditions. Darkfield microscopy has demonstrated the most limited sensitivity of the discussed techniques, mainly due to the D6 scaling of signal with particle size, which necessitates extremely efficient background rejection and extensive dynamic range to detect very small signals. Even in the absence of demonstrating single-molecule sensitivity, darkfield microscopy has been used to detect single viruses71,112 and 10 nm GNPs42 in studies on custom-built microscopes.
Despite the limited sensitivity of darkfield microscopy relative to interferometric scattering and plasmonic enhancement, efficient background rejection enables much higher illumination intensities for a given detector, allowing a significant portion of the full well depth of the camera to be used for detecting scattered photons. The combination of strong background rejection with a theoretically unlimited photon budget enables darkfield microscopy to perform well as a single-particle tracking technique when imaging suitably sized labels due to the high resulting signal-to-background ratios. Examples include shot-noise-limited localization precision of ∼1 nm with 10 μs temporal resolution when tracking 20 nm GNPs in a micromirror TIRS setup58 and 1.3 Å localization precision with 1 ms temporal resolution when tracking 40 nm GNPs using a perforated mirror TIRS setup with annular illumination.72
A potential downside of the single-particle tracking ability of darkfield microscopy is the difficulty of applying the technique to biological environments that exhibit high levels of background scattering. To overcome this limitation, strategies may include improved image processing techniques and the introduction of next-generation detectors with higher full-well capacities. We anticipate darkfield microscopy usage to continue to focus on single-particle tracking of biologically relevant systems using scattering labels, and expect that further technological developments will continue to push the boundaries of spatiotemporal resolution, enabling the analysis of even faster and more subtle molecular motion. Beyond this application, for darkfield microscopy to reach the sensitivity limits achieved by interferometric imaging, developments in image processing might be required to identify the target signal from any unwanted background. This may be a nontrivial task, however, as background scattering from sources near the target particle, such as coverslip surface roughness, may interfere with the scattered light from the particle, producing a complex signal. Thus, the ultimate challenge for darkfield microscopy, when attempting to observe very small scatterers such as single molecules, is to retain a purely scattering intensity measurement in the absence of any background, or to develop novel image analysis approaches, which allows the interferometric interaction of the signal and background to be used.
Interferometric scattering microscopy has exhibited sensitivity superior to that of darkfield microscopy, largely due to the weaker D3 scaling of the signal with particle size and resulting higher ease of background suppression. The boundaries have been significantly pushed, from the detection of metal nanoparticles to single proteins, through the introduction of innovative image processing, and data analysis techniques combined with various technological developments.32−34,43 Significantly improving the sensitivity of interferometric microscopy in its current form may prove a difficult task, as there are limits to the incident laser power that biological samples and optical elements can be exposed to. Strategies to collect more light by integrating for longer in time, at the expense of temporal resolution, would require an improved approach to background noise subtraction, especially in the context of lateral drift and other cumulative noise sources. This could be achieved by developments in both the hardware, such as substrate modifications, or the image analysis methods. Additionally, exploring mechanisms which enable an increase in the light–matter interaction strength at the sample might be worthwhile.
The ability to detect and quantify the mass of single proteins in solution under ambient conditions is an exciting development, with broad use across the life sciences. We anticipate the detection limit of MP to be further improved through a range of hardware and software developments, enabling the study of currently inaccessible systems and more complex biomolecular processes. iSCAT, one of the most rapidly developing interferometric scattering approaches, has transitioned from single-particle tracking in low scattering environments to 3D tracking in complex environments, including cell membranes.135−137,139 This represents an important advance, as tracking biomolecules within their native environments at spatiotemporal resolutions inaccessible in fluorescence microscopy paves the way to discover hitherto hidden dynamics. We hypothesize that interferometric scattering microscopy development will continue to focus on unlocking the information contained within its complex PSF, while improvements in sensitivity and image processing will enable the use of smaller scattering labels and imaging at higher spatiotemporal resolutions. In addition to pushing the boundaries of sensitivity, we expect interferometric scattering microscopy techniques to achieve some degree of specificity, for example, through the development of improved biosensor assays and technology, yielding ever-lower limits of detection.200,202,209
Similar to darkfield microscopy, interferometric scattering microscopy is well-suited to being combined with other microscopy techniques due to the simplicity of the optical setup and the universality of light scattering. In this review, we have discussed the incorporation of iSCAT detection in optical traps,138,144−148 however, this is just one of many techniques with which iSCAT is compatible. Therefore, we anticipate more correlative combinations to be explored, including combinations involving scanning probe and light microscopy techniques.
Further applications of iSCAT, some of which stretch beyond the scope of this review, focus on nanoscale imaging. Most recently, the concept of iSCAT’s general sensitivity to changes to local polarizability has been extended beyond the strict life sciences context. One example is the introduction of time-resolved iSCAT, termed stroboSCAT, where iSCAT is performed in a pump–probe configuration to monitor ultrafast dynamics in 2D materials following photoexcitation.309,310 iSCAT has also recently been used to monitor phase transitions during nanoparticle charging in batteries at the single-particle level, enabling detailed investigations of the process.311 In addition, another recent development is the combination of iSCAT detection principles with mid-infrared photothermal (MIP) microscopy. This emerging technique is particularly exciting due to the chemical specificity obtained during the measurement. In visible probe MIP microscopy, the mid-IR absorption induced photothermal effect is probed using visible light.312−314 The technique has recently been applied to chemically specific imaging of cells,315 100 nm polystyrene beads,316 and single virus particles.317
Significant advances have been made in surface plasmon resonance microscopy, the part of the plasmonic imaging field covered in this review. Developments in both the experimental methods and image analysis have enabled its application to the study of many biological molecules and systems, including viruses, DNA molecules, and single proteins, as well as larger complexes such as bacteria and live mammalian cells. Despite the near-field enhancement provided by the surface plasmon coupling of the incident light, the major limitation of SPRM is its complex, elongated PSF, which renders image contrast comparisons to nonplasmonically enhanced interferometric approaches difficult to elucidate. The lateral resolution, in the direction of surface plasmon propagation, is limited by the characteristic parabolic PSF tail, which extends for several micrometers beyond the diffraction limit of the transverse direction. Recent attempts to tackle this issue have shown promising results. Altering the optical system to generate images from multiple illumination directions has proven to be an effective approach,298 although it limits the temporal resolution (which is restricted to the beam-scanning rate) as well as the PSF (which remains suboptimal due to polarization effects and anisotropic emission). Novel image processing methods have aided the analysis of the SPRM PSF and also enabled the reconstruction of a symmetrical, diffraction limited PSF.307,308 The initial promise of these resolution-enhanced SPRM techniques bodes well for future iterations, which minimize any limitations associated with temporal resolution and image contrast.
Ever-expanding developments of the plasmonically active substrate, including the addition of bespoke nanostructures and nanoapertures, are enabling further near-field enhancement, as well as other experimental advantages, such as alternative optical options and specific surface functionalization. Bloch surface wave microscopy enables many of these advantages by replacing the metallic film with a dielectric multilayer, which may help to alleviate any negative effects associated with the heating of the metallic film. Specific parameters of this multilayer structure can be modified to suit a range of illumination wavelengths and the use of s-polarized light, while the SiO2 top layer resembles that of a typical microscope coverslip.298 Finally, the incorporation of surface-charge-sensitive measurements has shown great promise for improving SPRM sensitivity toward the detection of small molecules.288 We anticipate many more promising technical developments and novel applications introduced to the expanding field of plasmonic imaging in the coming years.
It is worth emphasizing the degree to which nominally very different methods share very similar challenges. Traditionally, darkfield, interferometric, and plasmonic microscopy have been largely separate fields. Given the advances in these fields, the next step changes likely require the combination of concepts and approaches from all three, together with stronger efforts in terms of optics, detectors, and data analysis geared toward label-free optical images. Concerted efforts have enabled revolutions in fluorescence and electron microscopy; they are now required to fulfill the promise that scattering-based methods have demonstrated over the past few decades.
Acknowledgments
P. K. is supported by an ERC Consolidator grant (PHOTOMASS 819593) and an EPSRC Leadership Fellowship (EP/T03419X/1). L.P. is supported by the Christ Church Moritz-Heyman Award and the EPSRC Doctoral Training Partnership. J.S.P. is supported by the EPSRC Doctoral Training Partnership. We thank Catherine Lichten and other members of the Kukura Group who provided highly valued feedback and input to this article. The field of optical microscopy, and its application to nanoscale phenomena has seen tremendous activity over the past decades, and while we have made our best effort in being comprehensive, it is likely unavoidable that we may have missed important and relevant contributions, for which we would like to apologize. We very much acknowledge all those who contribute to this incredibly exciting and rapidly progressing field.
Glossary
Abbreviations
- AOD
acousto-optic deflector
- Apols
amphipols
- ABEL
anti-Brownian electrokinetic
- ATR
attenuated total reflection
- ARI
azimuthal rotation illumination
- BFP
back focal plane
- BSW
Bloch surface wave
- BSWM
Bloch surface wave microscopy
- BSA
bovine serum albumin
- COBRI
coherent bright-field microscopy
- CCMV
cowpea chlorotic mottle virus
- cryo-EM
cryo-electron microscopy
- DIC
differential interference contrast
- DLS
dynamic light scattering
- FBS
fetal bovine serum
- FOV
field of view
- FWHM
full width at half-maximum
- GNP
gold nanoparticle
- GNR
gold nanorod
- HBsAg
hepatitis B surface antigen
- IgA
human immunoglobulin A
- IgM
human immunoglobulin M
- ITO
indium tin oxide
- IRM
interference reflection microscopy
- iPM
interferometric plasmonic microscopy
- IRIS
interferometric reflectance imaging sensor
- iSCAT
interferometric scattering microscopy
- LED
light emitting diode
- LSPR
localized surface plasmon
- LRSPR
long-range surface plasmon resonance
- MP
mass photometry
- MS
mass spectrometry
- MIP
mid-infrared photothermal
- nanoCET
nanocapillary electrophoretic tracking
- NA
numerical aperture
- OmpF
outer membrane protein
- PCM
phase contrast microscopy
- PT-SLM
photothermal spatial light modulator
- PFU
plaque-forming units
- PSM
plasmonic scattering microscopy
- P-EIM
plasmonic-based electrochemical impedance microscopy
- PSF
point-spread function
- PBS
polarizing beamsplitter
- PEG
polyethylene glycol
- QPD
quadrant photodiode
- QWP
quarter wave plate
- RICM
reflection interference contrast microscopy
- ROI
region of interest
- RF-SCAT
remote focusing interferometric scattering microscopy
- ROCS
rotating coherent scattering
- SEM
scanning electron microscopy
- SLSPM
scanning localized surface plasmon microscopy
- SNR
signal-to-noise ratio
- SP-IRIS
single-particle interferometric reflectance imaging sensor
- SPT
single-particle tracking
- miRNA
single-strand microRNA
- SP
surface plasmon
- SPP
surface plasmon polaritons
- SPR
surface plasmon resonance
- SPRI
surface plasmon resonance imaging
- SPRM
surface plasmon resonance microscopy
- SPCEM
surface plasmon-coupled emission microscopy
- TIR
total internal reflection
- TIRF
total internal reflection fluorescence
- TIRS
total internal reflection scattering
- 2D-SPR
two-dimensional surface plasmon resonance
- TFP
type IV pili
- VSV
vesicular stomatitis virus
- WSM
waveguide scattering microscopy
Biographies
Lee Priest studied Natural Sciences at University College London and received his MSci degree in 2016. He is currently a Ph.D. student in the Kukura Group at the University of Oxford. His research has focused on advancing the sensitivity of interferometric imaging techniques, primarily in the context of mass photometry particle detection.
Jack S. Peters studied at the University of Warwick and received his MChem degree in 2018. He is currently a graduate student under the supervision of Professor Philipp Kukura at the University of Oxford. His current research focuses on technological development of mass photometry, with the aim of improving the sensitivity and mass resolution of the technique.
Philipp Kukura is Professor of Chemistry and Fellow of Exeter College at the University of Oxford. He received an MChem from the University of Oxford (2002) and a Ph.D. from the University of California, Berkeley (2006). After postdoctoral work at ETH Zurich, he joined the Chemistry Department at the University of Oxford as Research Fellow (2010), Lecturer (2011), and then full Professor (2016). Honours and awards include the Royal Society of Chemistry Harrison Meldola Award (2011), the Royal Society of Chemistry Marlow Award (2015), the EBSA Young Investigator Award (2017), the Royal Society Wolfson Research Merit Award (2018), the Klung-Wilhelmy Science award in Chemistry (2018), the Blavatnik Award for Young Scientists UK in Chemistry (2019), and the RMS Medal for Light Microscopy (2021). His current research interests include the application of ultrasensitive light microscopy to study bimolecular structure and interactions.
Author Contributions
† L.P. and J.S.P. contributed equally.
The authors declare no competing financial interest.
References
- Binnig G.; Rohrer H.; Gerber C.; Weibel E. Surface Studies by Scanning Tunneling Microscopy. Phys. Rev. Lett. 1982, 49, 57–61. 10.1103/PhysRevLett.49.57. [DOI] [Google Scholar]
- Eigler D. M.; Schweizer E. K. Positioning Single Atoms with a Scanning Tunnelling Microscope. Nature 1990, 344, 524–526. 10.1038/344524a0. [DOI] [Google Scholar]
- Binnig G.; Quate C. F.; Gerber C. Atomic Force Microscope. Phys. Rev. Lett. 1986, 56, 930–933. 10.1103/PhysRevLett.56.930. [DOI] [PubMed] [Google Scholar]
- Ando T.; Kodera N.; Takai E.; Maruyama D.; Saito K.; Toda A. A High-Speed Atomic Force Microscope for Studying Biological Macromolecules. Proc. Natl. Acad. Sci. U. S. A. 2001, 98, 12468–12472. 10.1073/pnas.211400898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moerner W. E.; Kador L. Optical Detection and Spectroscopy of Single Molecules in a Solid. Phys. Rev. Lett. 1989, 62, 2535–2538. 10.1103/PhysRevLett.62.2535. [DOI] [PubMed] [Google Scholar]
- Orrit M.; Bernard J. Single Pentacene Molecules Detected by Fluorescence Excitation in a P-Terphenyl Crystal. Phys. Rev. Lett. 1990, 65, 2716–2719. 10.1103/PhysRevLett.65.2716. [DOI] [PubMed] [Google Scholar]
- Neuhauser W.; Hohenstatt M.; Toschek P. E.; Dehmelt H. Localized Visible Ba+ Mono-Ion Oscillator. Phys. Rev. A: At., Mol., Opt. Phys. 1980, 22, 1137–1140. 10.1103/PhysRevA.22.1137. [DOI] [Google Scholar]
- Moerner W. E.; Orrit M. Illuminating Single Molecules in Condensed Matter. Science 1999, 283, 1670–1676. 10.1126/science.283.5408.1670. [DOI] [PubMed] [Google Scholar]
- Dickson R. M.; Cubitt A. B.; Tsien R. Y.; Moerner W. E. On/off Blinking and Switching Behaviour of Single Molecules of Green Fluorescent Protein. Nature 1997, 388, 355–358. 10.1038/41048. [DOI] [PubMed] [Google Scholar]
- Betzig E.; Patterson G. H.; Sougrat R.; Lindwasser O. W.; Olenych S.; Bonifacino J. S.; Davidson M. W.; Lippincott-Schwartz J.; Hess H. F. Imaging Intracellular Fluorescent Proteins at Nanometer Resolution. Science 2006, 313, 1642–1645. 10.1126/science.1127344. [DOI] [PubMed] [Google Scholar]
- Rust M. J.; Bates M.; Zhuang X. Sub-Diffraction-Limit Imaging by Stochastic Optical Reconstruction Microscopy (STORM). Nat. Methods 2006, 3, 793–795. 10.1038/nmeth929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hess S. T.; Girirajan T. P. K.; Mason M. D. Ultra-High Resolution Imaging by Fluorescence Photoactivation Localization Microscopy. Biophys. J. 2006, 91, 4258–4272. 10.1529/biophysj.106.091116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dempsey G. T.; Vaughan J. C.; Chen K. H.; Bates M.; Zhuang X. Evaluation of Fluorophores for Optimal Performance in Localization-Based Super-Resolution Imaging. Nat. Methods 2011, 8, 1027–1040. 10.1038/nmeth.1768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gwosch K. C.; Pape J. K.; Balzarotti F.; Hoess P.; Ellenberg J.; Ries J.; Hell S. W. MINFLUX Nanoscopy Delivers 3D Multicolor Nanometer Resolution in Cells. Nat. Methods 2020, 17, 217–224. 10.1038/s41592-019-0688-0. [DOI] [PubMed] [Google Scholar]
- Siedentopf H.; Zsigmondy R. Uber Sichtbarmachung Und Größenbestimmung Ultramikoskopischer Teilchen, Mit Besonderer Anwendung Auf Goldrubingläser. Ann. Phys. 1902, 315, 1–39. 10.1002/andp.19023150102. [DOI] [Google Scholar]
- Zernike F. Das Phasenkontrastverfahren Bei Der Mikroskopischen Beobachtung. Z. Technol. Phys. 1935, 16, 454–457. [Google Scholar]
- Zernike F. Phase Contrast, a New Method for the Microscopic Observation of Transparent Objects. Physica 1942, 9, 686–698. 10.1016/S0031-8914(42)80035-X. [DOI] [Google Scholar]
- Zernike F. Phase Contrast, a New Method for the Microscopic Observation of Transparent Objects Part II. Physica 1942, 9, 974–986. 10.1016/S0031-8914(42)80079-8. [DOI] [Google Scholar]
- Nomarski G. Microinterféromètre Différentiel à Ondes Polarisées. J. Phys. Radium 1955, 16, 9S–11S. [Google Scholar]
- Allen R. D.; David G. B.; Nomarski G. The Zeiss-Nomarski Differential Interference Equipment for Transmitted-Light Microscopy. Z. Wiss. Mikrosk. 1969, 69, 193–221. [PubMed] [Google Scholar]
- Curtis A. S. The Mechanism of Adhesion of Cells to Glass. A Study by Interference Reflection Microscopy. J. Cell Biol. 1964, 20, 199–215. 10.1083/jcb.20.2.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verschueren H. Interference Reflection Microscopy in Cell Biology: Methodology and Applications. J. Cell Sci. 1985, 75, 279–301. 10.1242/jcs.75.1.279. [DOI] [PubMed] [Google Scholar]
- Ploem J. S.Reflection-Contrast Microscopy as a Tool for Investigation of the Attachment of Living Cells to a Glass Surface; Van Furth R., Ed.; Blackwell Scientific: Oxford, 1975. [Google Scholar]
- Limozin L.; Sengupta K. Quantitative Reflection Interference Contrast Microscopy (RICM) in Soft Matter and Cell Adhesion. ChemPhysChem 2009, 10, 2752–2768. 10.1002/cphc.200900601. [DOI] [PubMed] [Google Scholar]
- Gaiduk A.; Yorulmaz M.; Ruijgrok P. V.; Orrit M. Room-Temperature Detection of a Single Molecule’s Absorption by Photothermal Contrast. Science 2010, 330, 353–356. 10.1126/science.1195475. [DOI] [PubMed] [Google Scholar]
- Celebrano M.; Kukura P.; Renn A.; Sandoghdar V. Single-Molecule Imaging by Optical Absorption. Nat. Photonics 2011, 5, 95–98. 10.1038/nphoton.2010.290. [DOI] [Google Scholar]
- Chong S.; Min W.; Xie X. S. Ground-State Depletion Microscopy: Detection Sensitivity of Single-Molecule Optical Absorption at Room Temperature. J. Phys. Chem. Lett. 2010, 1, 3316–3322. 10.1021/jz1014289. [DOI] [Google Scholar]
- Vollmer F.; Arnold S. Whispering-Gallery-Mode Biosensing: Label-Free Detection down to Single Molecules. Nat. Methods 2008, 5, 591–596. 10.1038/nmeth.1221. [DOI] [PubMed] [Google Scholar]
- Vollmer F.; Swaim J. D.; Foreman M. R. Whispering Gallery Mode Sensors. Adv. Opt. Photonics 2015, 7, 168–240. 10.1364/AOP.7.000168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baaske M. D.; Vollmer F. Optical Observation of Single Atomic Ions Interacting with Plasmonic Nanorods in Aqueous Solution. Nat. Photonics 2016, 10, 733–739. 10.1038/nphoton.2016.177. [DOI] [Google Scholar]
- Abbe E. Beiträge Zur Theorie Des Mikroskops Und Der Mikroskopischen Wahrnehmung: I. Die Construction von Mikroskopen Auf Grund Der Theorie. Arch. für mikroskopische Anat. 1873, 9, 413–418. 10.1007/BF02956173. [DOI] [Google Scholar]
- Ortega Arroyo J.; Andrecka J.; Spillane K. M.; Billington N.; Takagi Y.; Sellers J. R.; Kukura P. Label-Free, All-Optical Detection, Imaging, and Tracking of a Single Protein. Nano Lett. 2014, 14, 2065–2070. 10.1021/nl500234t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piliarik M.; Sandoghdar V. Direct Optical Sensing of Single Unlabelled Proteins and Super-Resolution Imaging of Their Binding Sites. Nat. Commun. 2014, 5, 4495. 10.1038/ncomms5495. [DOI] [PubMed] [Google Scholar]
- Young G.; Hundt N.; Cole D.; Fineberg A.; Andrecka J.; Tyler A.; Olerinyova A.; Ansari A.; Marklund E. G.; Collier M. P.; et al. Quantitative Mass Imaging of Single Biological Macromolecules. Science 2018, 360, 423–427. 10.1126/science.aar5839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stallinga S.; Rieger B. Accuracy of the Gaussian Point Spread Function Model in 2D Localization Microscopy. Opt. Express 2010, 18, 24461. 10.1364/OE.18.024461. [DOI] [PubMed] [Google Scholar]
- Thompson R. E.; Larson D. R.; Webb W. W. Precise Nanometer Localization Analysis for Individual Fluorescent Probes. Biophys. J. 2002, 82, 2775–2783. 10.1016/S0006-3495(02)75618-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bohren C. F.; Huffman D. R.. Absorption and Scattering of Light by Small Particles; John Wiley & Sons: New York, 1983. [Google Scholar]
- Agio M.; Mojarad N. M.; Sandoghdar V. Plasmon Spectra of Nanospheres under a Tightly Focused Beam. J. Opt. Soc. Am. B 2008, 25, 651–658. 10.1364/JOSAB.25.000651. [DOI] [Google Scholar]
- Lieb M. A.; Zavislan J. M.; Novotny L. Single-Molecule Orientations Determined by Direct Emission Pattern Imaging. J. Opt. Soc. Am. B 2004, 21, 1210-1215. 10.1364/JOSAB.21.001210. [DOI] [Google Scholar]
- Brokmann X.; Coolen L.; Hermier J. P.; Dahan M. Emission Properties of Single CdSe/ZnS Quantum Dots Close to a Dielectric Interface. Chem. Phys. 2005, 318, 91–98. 10.1016/j.chemphys.2005.06.032. [DOI] [Google Scholar]
- Lee K. G.; Chen X. W.; Eghlidi H.; Kukura P.; Lettow R.; Renn A.; Sandoghdar V.; Götzinger S. A Planar Dielectric Antenna for Directional Single-Photon Emission and near-Unity Collection Efficiency. Nature Photonics 2011, 166–169. 10.1038/nphoton.2010.312. [DOI] [Google Scholar]
- Weigel A.; Sebesta A.; Kukura P. Dark Field Microspectroscopy with Single Molecule Fluorescence Sensitivity. ACS Photonics 2014, 1, 848–856. 10.1021/ph500138u. [DOI] [Google Scholar]
- Cole D.; Young G.; Weigel A.; Sebesta A.; Kukura P. Label-Free Single-Molecule Imaging with Numerical-Aperture-Shaped Interferometric Scattering Microscopy. ACS Photonics 2017, 4, 211–216. 10.1021/acsphotonics.6b00912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liebel M.; Hugall J. T.; Van Hulst N. F. Ultrasensitive Label-Free Nanosensing and High-Speed Tracking of Single Proteins. Nano Lett. 2017, 17, 1277–1281. 10.1021/acs.nanolett.6b05040. [DOI] [PubMed] [Google Scholar]
- Avci O.; Campana M. I.; Yurdakul C.; Selim Ünlü M. Pupil Function Engineering for Enhanced Nanoparticle Visibility in Wide-Field Interferometric Microscopy. Optica 2017, 4, 247-254. 10.1364/OPTICA.4.000247. [DOI] [Google Scholar]
- Horio T.; Hotani H. Visualization of the Dynamic Instability of Individual Microtubules by Dark-Field Microscopy. Nature 1986, 321, 605–607. 10.1038/321605a0. [DOI] [PubMed] [Google Scholar]
- Kudo S.; Magariyama Y.; Aizawa S. I. Abrupt Changes in Flagellar Rotation Observed by Laser Dark-Field Microscopy. Nature 1990, 346, 677–680. 10.1038/346677a0. [DOI] [PubMed] [Google Scholar]
- Nishiyama M.; Muto E.; Inoue Y.; Yanagida T.; Higuchi H. Substeps within the 8-Nm Step of the ATPase Cycle of Single Kinesin Molecules. Nat. Cell Biol. 2001, 3, 425–428. 10.1038/35070116. [DOI] [PubMed] [Google Scholar]
- Yasuda R.; Noji H.; Yoshida M.; Kinosita K.; Itoh H. Resolution of Distinct Rotational Substeps by Submillisecond Kinetic Analysis of F1-ATPase. Nature 2001, 410, 898–904. 10.1038/35073513. [DOI] [PubMed] [Google Scholar]
- Dunn A. R.; Spudich J. A. Dynamics of the Unbound Head during Myosin V Processive Translocation. Nat. Struct. Mol. Biol. 2007, 14, 246–248. 10.1038/nsmb1206. [DOI] [PubMed] [Google Scholar]
- Axelrod D. Total Internal Reflection Fluorescence Microscopy in Cell Biology. Traffic 2001, 2, 764–774. 10.1034/j.1600-0854.2001.21104.x. [DOI] [PubMed] [Google Scholar]
- Schneckenburger H. Total Internal Reflection Fluorescence Microscopy: Technical Innovations and Novel Applications. Curr. Opin. Biotechnol. 2005, 16, 13–18. 10.1016/j.copbio.2004.12.004. [DOI] [PubMed] [Google Scholar]
- Mattheyses A. L.; Simon S. M.; Rappoport J. Z. Imaging with Total Internal Reflection Fluorescence Microscopy for the Cell Biologist. J. Cell Sci. 2010, 123, 3621–3628. 10.1242/jcs.056218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ambrose E. J. A Surface Contact Microscope for the Study of Cell Movements. Nature 1956, 178, 1194. 10.1038/1781194a0. [DOI] [PubMed] [Google Scholar]
- Ambrose E. J. The Movements of Fibrocytes. Exp. Cell Res. 1961, 8, 54–73. 10.1016/0014-4827(61)90340-8. [DOI] [PubMed] [Google Scholar]
- Braslavsky I.; Amit R.; Jaffar Ali B. M.; Gileadi O.; Oppenheim A.; Stavans J. Objective-Type Dark-Field Illumination for Scattering from Microbeads. Appl. Opt. 2001, 40, 5650–5657. 10.1364/AO.40.005650. [DOI] [PubMed] [Google Scholar]
- Stout A. L.; Axelrod D. Evanescent Field Excitation of Fluorescence by Epi-Illumination Microscopy. Appl. Opt. 1989, 28, 5237–5242. 10.1364/AO.28.005237. [DOI] [PubMed] [Google Scholar]
- Meng X.; Sonn-Segev A.; Schumacher A.; Cole D.; Young G.; Thorpe S.; Style R. W.; Dufresne E. R.; Kukura P.. Micromirror Total Internal Reflection Microscopy for High-Performance Single Particle Tracking at Interfaces. arXiv 2021, 2103.09738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ueno H.; Nishikawa S.; Iino R.; Tabata K. V.; Sakakihara S.; Yanagida T.; Noji H. Simple Dark-Field Microscopy with Nanometer Spatial Precision and Microsecond Temporal Resolution. Biophys. J. 2010, 98, 2014–2023. 10.1016/j.bpj.2010.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kobayashi R.; Ueno H.; Li C. B.; Noji H. Rotary Catalysis of Bovine Mitochondrial F1-ATPase Studied by Single-Molecule Experiments. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 1447–1456. 10.1073/pnas.1909407117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe R.; Minagawa Y.; Noji H. Thermodynamic Analysis of F 1 -ATPase Rotary Catalysis Using High-Speed Imaging. Protein Sci. 2014, 23, 1773–1779. 10.1002/pro.2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C. B.; Ueno H.; Watanabe R.; Noji H.; Komatsuzaki T. ATP Hydrolysis Assists Phosphate Release and Promotes Reaction Ordering in F1 -ATPase. Nat. Commun. 2015, 6, 10223. 10.1038/ncomms10223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe R.; Hayashi K.; Ueno H.; Noji H. Catalysis-Enhancement via Rotary Fluctuation of F1-Atpase. Biophys. J. 2013, 105, 2385–2391. 10.1016/j.bpj.2013.09.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayashi S.; Ueno H.; Shaikh A. R.; Umemura M.; Kamiya M.; Ito Y.; Ikeguchi M.; Komoriya Y.; Iino R.; Noji H. Molecular Mechanism of ATP Hydrolysis in F1-ATPase Revealed by Molecular Simulations and Single-Molecule Observations. J. Am. Chem. Soc. 2012, 134, 8447–8454. 10.1021/ja211027m. [DOI] [PubMed] [Google Scholar]
- Minagawa Y.; Ueno H.; Hara M.; Ishizuka-Katsura Y.; Ohsawa N.; Terada T.; Shirouzu M.; Yokoyama S.; Yamato I.; Muneyuki E.; et al. Basic Properties of Rotary Dynamics of the Molecular Motor Enterococcus Hirae V1-ATPase. J. Biol. Chem. 2013, 288, 32700–32707. 10.1074/jbc.M113.506329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iida T.; Minagawa Y.; Ueno H.; Kawai F.; Murata T.; Iino R. Single-Molecule Analysis Reveals Rotational Substeps and Chemo-Mechanical Coupling Scheme of Enterococcus Hirae V1-ATPase. J. Biol. Chem. 2019, 294, 17017–17030. 10.1074/jbc.RA119.008947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ueno H.; Minagawa Y.; Hara M.; Rahman S.; Yamato I.; Muneyuki E.; Noji H.; Murata T.; Iino R. Torque Generation of Enterococcus Hirae V-ATPase. J. Biol. Chem. 2014, 289, 31212–31223. 10.1074/jbc.M114.598177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nishikawa S.; Arimoto I.; Ikezaki K.; Sugawa M.; Ueno H.; Komori T.; Iwane A. H.; Yanagida T. Switch between Large Hand-Over-Hand and Small Inchworm-like Steps in Myosin VI. Cell 2010, 142, 879–888. 10.1016/j.cell.2010.08.033. [DOI] [PubMed] [Google Scholar]
- Isojima H.; Iino R.; Niitani Y.; Noji H.; Tomishige M. Direct Observation of Intermediate States during the Stepping Motion of Kinesin-1. Nat. Chem. Biol. 2016, 12, 290–297. 10.1038/nchembio.2028. [DOI] [PubMed] [Google Scholar]
- Nakamura A.; Okazaki K.-i; Furuta T.; Sakurai M.; Iino R. Processive Chitinase Is Brownian Monorail Operated by Fast Catalysis after Peeling Rail from Crystalline Chitin. Nat. Commun. 2018, 9, 3814. 10.1038/s41467-018-06362-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Enoki S.; Iino R.; Morone N.; Kaihatsu K.; Sakakihara S.; Kato N.; Noji H. Label-Free Single-Particle Imaging of the Influenza Virus by Objective-Type Total Internal Reflection Dark-Field Microscopy. PLoS One 2012, 7, e49208 10.1371/journal.pone.0049208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ando J.; Nakamura A.; Visootsat A.; Yamamoto M.; Song C.; Murata K.; Iino R. Single-Nanoparticle Tracking with Angstrom Localization Precision and Microsecond Time Resolution. Biophys. J. 2018, 115, 2413–2427. 10.1016/j.bpj.2018.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ando J.; Shima T.; Kanazawa R.; Shimo-Kon R.; Nakamura A.; Yamamoto M.; Kon T.; Iino R. Small Stepping Motion of Processive Dynein Revealed by Load-Free High-Speed Single-Particle Tracking. Sci. Rep. 2020, 10, 1080. 10.1038/s41598-020-58070-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ando J.; Nakamura A.; Yamamoto M.; Song C.; Murata K.; Iino R. Multicolor High-Speed Tracking of Single Biomolecules with Silver, Gold, and Silver-Gold Alloy Nanoparticles. ACS Photonics 2019, 6, 2870–2883. 10.1021/acsphotonics.9b00953. [DOI] [Google Scholar]
- Sönnichsen C.; Alivisatos A. P. Gold Nanorods as Novel Nonbleaching Plasmon-Based Orientation Sensors for Polarized Single-Particle Microscopy. Nano Lett. 2005, 5, 301–304. 10.1021/nl048089k. [DOI] [PubMed] [Google Scholar]
- Huang X.; Neretina S.; El-Sayed M. A. Gold Nanorods: From Synthesis and Properties to Biological and Biomedical Applications. Adv. Mater. 2009, 21, 4880–4910. 10.1002/adma.200802789. [DOI] [PubMed] [Google Scholar]
- Mazaheri M.; Ehrig J.; Shkarin A.; Zaburdaev V.; Sandoghdar V. Ultrahigh-Speed Imaging of Rotational Diffusion on a Lipid Bilayer. Nano Lett. 2020, 20, 7213–7219. 10.1021/acs.nanolett.0c02516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wicker K.; Heintzmann R. Resolving a Misconception about Structured Illumination. Nat. Photonics 2014, 8, 342–344. 10.1038/nphoton.2014.88. [DOI] [Google Scholar]
- Cronin D. J.; Smith A. E. Dynamic Coherent Optical System. Opt. Eng. 1973, 12, 120250. 10.1117/12.7971629. [DOI] [Google Scholar]
- Cotte Y.; Toy F.; Jourdain P.; Pavillon N.; Boss D.; Magistretti P.; Marquet P.; Depeursinge C. Marker-Free Phase Nanoscopy. Nat. Photonics 2013, 7, 113–117. 10.1038/nphoton.2012.329. [DOI] [Google Scholar]
- Zheng G.; Horstmeyer R.; Yang C. Wide-Field, High-Resolution Fourier Ptychographic Microscopy. Nat. Photonics 2013, 7, 739–745. 10.1038/nphoton.2013.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maire G.; Giovannini H.; Talneau A.; Chaumet P. C.; Belkebir K.; Sentenac A. Phase Imaging and Synthetic Aperture Super-Resolution via Total Internal Reflection Microscopy. Opt. Lett. 2018, 43, 2173–2176. 10.1364/OL.43.002173. [DOI] [PubMed] [Google Scholar]
- Chowdhury S.; Dhalla A.-H.; Izatt J. Structured Oblique Illumination Microscopy for Enhanced Resolution Imaging of Non-Fluorescent, Coherently Scattering Samples. Biomed. Opt. Express 2012, 3, 1841–1854. 10.1364/BOE.3.001841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- von Olshausen P.; Rohrbach A. Coherent Total Internal Reflection Dark-Field Microscopy: Label-Free Imaging beyond the Diffraction Limit. Opt. Lett. 2013, 38, 4066–4069. 10.1364/OL.38.004066. [DOI] [PubMed] [Google Scholar]
- Jünger F.; Olshausen P. V.; Rohrbach A. Fast, Label-Free Super-Resolution Live-Cell Imaging Using Rotating Coherent Scattering (ROCS) Microscopy. Sci. Rep. 2016, 6, 30393. 10.1038/srep30393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heintzmann R.; Cremer C. G.. Laterally Modulated Excitation Microscopy: Improvement of Resolution by Using a Diffraction Grating. In Optical Biopsies and Microscopic Techniques III; SPIE, 1999; Vol. 3568, pp 185–196 10.1117/12.336833. [DOI] [Google Scholar]
- Gustafsson M. G. L. Surpassing the Lateral Resolution Limit by a Factor of Two Using Structured Illumination Microscopy. J. Microsc. 2000, 198, 82–87. 10.1046/j.1365-2818.2000.00710.x. [DOI] [PubMed] [Google Scholar]
- Heintzmann R.; Huser T. Super-Resolution Structured Illumination Microscopy. Chem. Rev. 2017, 117, 13890–13908. 10.1021/acs.chemrev.7b00218. [DOI] [PubMed] [Google Scholar]
- Koch M. D.; Rohrbach A. Label-Free Imaging and Bending Analysis of Microtubules by ROCS Microscopy and Optical Trapping. Biophys. J. 2018, 114, 168–177. 10.1016/j.bpj.2017.10.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jünger F.; Rohrbach A. Strong Cytoskeleton Activity on Millisecond Timescales upon Particle Binding Revealed by ROCS Microscopy. Cytoskeleton 2018, 75, 410–424. 10.1002/cm.21478. [DOI] [PubMed] [Google Scholar]
- Ruh D.; Mutschler J.; Michelbach M.; Rohrbach A. Superior Contrast and Resolution by Image Formation in Rotating Coherent Scattering (ROCS) Microscopy. Optica 2018, 5, 1371–1381. 10.1364/OPTICA.5.001371. [DOI] [Google Scholar]
- Nan X.; Sims P. A.; Xie X. S. Organelle Tracking in a Living Cell with Microsecond Time Resolution and Nanometer Spatial Precision. ChemPhysChem 2008, 9, 707–712. 10.1002/cphc.200700839. [DOI] [PubMed] [Google Scholar]
- Sudhakar S.; Abdosamadi M. K.; Jachowski T. J.; Bugiel M.; Jannasch A.; Schäffer E. Germanium Nanospheres for Ultraresolution Picotensiometry of Kinesin Motors. Science 2021, 371, eabd9944 10.1126/science.abd9944. [DOI] [PubMed] [Google Scholar]
- Mickolajczyk K. J.; Deffenbaugh N. C.; Ortega Arroyo J.; Andrecka J.; Kukura P.; Hancock W. O. Kinetics of Nucleotide-Dependent Structural Transitions in the Kinesin-1 Hydrolysis Cycle. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, E7186–E7193. 10.1073/pnas.1517638112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sowa Y.; Steel B. C.; Berry R. M. A Simple Backscattering Microscope for Fast Tracking of Biological Molecules. Rev. Sci. Instrum. 2010, 81, 113704. 10.1063/1.3495960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nord A. L.; Sowa Y.; Steel B. C.; Lo C. J.; Berry R. M. Speed of the Bacterial Flagellar Motor near Zero Load Depends on the Number of Stator Units. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, 11603–11608. 10.1073/pnas.1708054114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lo C. J.; Sowa Y.; Pilizota T.; Berry R. M. Mechanism and Kinetics of a Sodium-Driven Bacterial Flagellar Motor. Proc. Natl. Acad. Sci. U. S. A. 2013, 110, E2544–E2551. 10.1073/pnas.1301664110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao L.; Qiao Y.; He Y.; Yeung E. S. Three Dimensional Orientational Imaging of Nanoparticles with Darkfield Microscopy. Anal. Chem. 2010, 82, 5268–5274. 10.1021/ac1006848. [DOI] [PubMed] [Google Scholar]
- Enoki S.; Iino R.; Niitani Y.; Minagawa Y.; Tomishige M.; Noji H. High-Speed Angle-Resolved Imaging of a Single Gold Nanorod with Microsecond Temporal Resolution and One-Degree Angle Precision. Anal. Chem. 2015, 87, 2079–2086. 10.1021/ac502408c. [DOI] [PubMed] [Google Scholar]
- Grandin H. M.; Städler B.; Textor M.; Vörös J. Waveguide Excitation Fluorescence Microscopy: A New Tool for Sensing and Imaging the Biointerface. Biosens. Bioelectron. 2006, 21, 1476–1482. 10.1016/j.bios.2005.06.011. [DOI] [PubMed] [Google Scholar]
- Agnarsson B.; Ingthorsson S.; Gudjonsson T.; Leosson K. Evanescent-Wave Fluorescence Microscopy Using Symmetric Planar Waveguides. Opt. Express 2009, 17, 5075–5082. 10.1364/OE.17.005075. [DOI] [PubMed] [Google Scholar]
- Archetti A.; Glushkov E.; Sieben C.; Stroganov A.; Radenovic A.; Manley S. Waveguide-PAINT Offers an Open Platform for Large Field-of-View Super-Resolution Imaging. Nat. Commun. 2019, 10, 1267. 10.1038/s41467-019-09247-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thoma F.; Langbein U.; Mittler-Neher S. Waveguide Scattering Microscopy. Opt. Commun. 1997, 134, 16–20. 10.1016/S0030-4018(96)00535-4. [DOI] [Google Scholar]
- Ni W.; Chen H.; Kou X.; Yeung M. H.; Wang J. Optical Fiber-Excited Surface Plasmon Resonance Spectroscopy of Single and Ensemble Gold Nanorods. J. Phys. Chem. C 2008, 112, 8105–8109. 10.1021/jp801579m. [DOI] [Google Scholar]
- Hill D. J.; Pinion C. W.; Christesen J. D.; Cahoon J. F. Waveguide Scattering Microscopy for Dark-Field Imaging and Spectroscopy of Photonic Nanostructures. ACS Photonics 2014, 1, 725–731. 10.1021/ph5001617. [DOI] [Google Scholar]
- Agnarsson B.; Lundgren A.; Gunnarsson A.; Rabe M.; Kunze A.; Mapar M.; Simonsson L.; Bally M.; Zhdanov V. P.; Höök F. Evanescent Light-Scattering Microscopy for Label-Free Interfacial Imaging: From Single Sub-100 Nm Vesicles to Live Cells. ACS Nano 2015, 9, 11849–11862. 10.1021/acsnano.5b04168. [DOI] [PubMed] [Google Scholar]
- Agnarsson B.; Mapar M.; Sjöberg M.; Alizadehheidari M.; Höök F. Low-Temperature Fabrication and Characterization of a Symmetric Hybrid Organic-Inorganic Slab Waveguide for Evanescent Light Microscopy. Nano Futures 2018, 2, 025007. 10.1088/2399-1984/aac25d. [DOI] [Google Scholar]
- Lundgren A.; Agnarsson rn; Zirbs R.; Zhdanov V. P.; Reimhult E.; Hook F. Nonspecific Colloidal-Type Interaction Explains Size-Dependent Specific Binding of Membrane-Targeted Nanoparticles Article. ACS Nano 2016, 10, 9974–9982. 10.1021/acsnano.6b04160. [DOI] [PubMed] [Google Scholar]
- Agnarsson B.; Wayment-Steele H. K.; Höök F.; Kunze A. Monitoring of Single and Double Lipid Membrane Formation with High Spatiotemporal Resolution Using Evanescent Light Scattering Microscopy †. Nanoscale 2016, 8, 19219. 10.1039/C6NR06726C. [DOI] [PubMed] [Google Scholar]
- Rupert D. L. M.; Mapar M.; Shelke G. V.; Norling K.; Elmeskog M.; Lotvall J. O.; Block S.; Bally M.; Agnarsson rn; Hook F. Effective Refractive Index and Lipid Content of Extracellular Vesicles Revealed Using Optical Waveguide Scattering and Fluorescence Microscopy. Langmuir 2018, 34, 8522–8531. 10.1021/acs.langmuir.7b04214. [DOI] [PubMed] [Google Scholar]
- Hannestad J. K.; Rocha S.; Agnarsson B.; Zhdanov V. P.; Wittung-Stafshede P.; Höök F. Single-Vesicle Imaging Reveals Lipid-Selective and Stepwise Membrane Disruption by Monomeric α-Synuclein. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 14178–14186. 10.1073/pnas.1914670117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faez S.; Lahini Y.; Weidlich S.; Garmann R. F.; Wondraczek K.; Zeisberger M.; Schmidt M. A.; Orrit M.; Manoharan V. N. Fast, Label-Free Tracking of Single Viruses and Weakly Scattering Nanoparticles in a Nanofluidic Optical Fiber. ACS Nano 2015, 9, 12349–12357. 10.1021/acsnano.5b05646. [DOI] [PubMed] [Google Scholar]
- Faez S.; Samin S.; Baasanjav D.; Weidlich S.; Schmidt M.; Mosk A. P. Nanocapillary Electrokinetic Tracking for Monitoring Charge Fluctuations on a Single Nanoparticle. Faraday Discuss. 2016, 193, 447–458. 10.1039/C6FD00097E. [DOI] [PubMed] [Google Scholar]
- Jiang S.; Zhao J.; Förster R.; Weidlich S.; Plidschun M.; Kobelke J.; Fatobene Ando R.; Schmidt M. A. Three Dimensional Spatiotemporal Nano-Scale Position Retrieval of the Confined Diffusion of Nano-Objects inside Optofluidic Microstructured Fibers. Nanoscale 2020, 12, 3146–3156. 10.1039/C9NR10351A. [DOI] [PubMed] [Google Scholar]
- Jiang S.; Förster R.; Plidschun M.; Kobelke J.; Fatobene Ando R.; Schmidt M. A. Three-Dimensional Spatiotemporal Tracking of Nano-Objects Diffusing in Water-Filled Optofluidic Microstructured Fiber. Nanophotonics 2020, 9, 4545–4554. 10.1515/nanoph-2020-0330. [DOI] [PubMed] [Google Scholar]
- Förster R.; Weidlich S.; Nissen M.; Wieduwilt T.; Kobelke J.; Goldfain A. M.; Chiang T. K.; Garmann R. F.; Manoharan V. N.; Lahini Y.; et al. Tracking and Analyzing the Brownian Motion of Nano-Objects Inside Hollow Core Fibers. ACS Sensors 2020, 5, 879–886. 10.1021/acssensors.0c00339. [DOI] [PubMed] [Google Scholar]
- Hartung A.; Kobelke J.; Schwuchow A.; Bierlich J.; Popp J.; Schmidt M. A.; Frosch T. Low-Loss Single-Mode Guidance in Large-Core Antiresonant Hollow-Core Fibers. Opt. Lett. 2015, 40, 3432–3435. 10.1364/OL.40.003432. [DOI] [PubMed] [Google Scholar]
- Kukura P.; Ewers H.; Müller C.; Renn A.; Helenius A.; Sandoghdar V. High-Speed Nanoscopic Tracking of the Position and Orientation of a Single Virus. Nat. Methods 2009, 6, 923–927. 10.1038/nmeth.1395. [DOI] [PubMed] [Google Scholar]
- Amos L. A.; Amos W. B. The Bending of Sliding Microtubules Imaged by Confocal Light Microscopy and Negative Stain Electron Microscopy. J. Cell Sci. 1991, 1991, 95–101. 10.1242/jcs.1991.Supplement_14.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindfors K.; Kalkbrenner T.; Stoller P.; Sandoghdar V. Detection and Spectroscopy of Gold Nanoparticles Using Supercontinuum White Light Confocal Microscopy. Phys. Rev. Lett. 2004, 93, 037401. 10.1103/PhysRevLett.93.037401. [DOI] [PubMed] [Google Scholar]
- Jacobsen V.; Stoller P.; Brunner C.; Vogel V.; Sandoghdar V. Interferometric Optical Detection and Tracking of Very Small Gold Nanoparticles at a Water-Glass Interface. Opt. Express 2006, 14, 405–414. 10.1364/OPEX.14.000405. [DOI] [PubMed] [Google Scholar]
- Ewers H.; Jacobsen V.; Klotzsch E.; Smith A. E.; Helenius A.; Sandoghdar V. Label-Free Optical Detection and Tracking of Single Virions Bound to Their Receptors in Supported Membrane Bilayers. Nano Lett. 2007, 7, 2263–2266. 10.1021/nl070766y. [DOI] [PubMed] [Google Scholar]
- Kukura P.; Celebrano M.; Renn A.; Sandoghdar V. Imaging a Single Quantum Dot When It Is Dark. Nano Lett. 2009, 9, 926–929. 10.1021/nl801735y. [DOI] [PubMed] [Google Scholar]
- Holanová K.; Vala M.; Piliarik M. Optical Imaging and Localization of Prospective Scattering Labels Smaller than a Single Protein. Opt. Laser Technol. 2019, 109, 323–327. 10.1016/j.optlastec.2018.08.014. [DOI] [Google Scholar]
- Ortega-Arroyo J.; Kukura P. Interferometric Scattering Microscopy (ISCAT): New Frontiers in Ultrafast and Ultrasensitive Optical Microscopy. Phys. Chem. Chem. Phys. 2012, 14, 15625–15636. 10.1039/c2cp41013c. [DOI] [PubMed] [Google Scholar]
- Taylor R. W.; Sandoghdar V. Interferometric Scattering Microscopy: Seeing Single Nanoparticles and Molecules via Rayleigh Scattering. Nano Lett. 2019, 19, 4827–4835. 10.1021/acs.nanolett.9b01822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor R. W.; Sandoghdar V.. Interferometric Scattering (ISCAT) Microscopy and Related Techniques. In Label-Free Super-Resolution Microscopy. Biological and Medical Physics, Biomedical Engineering; Astratov V., Ed.; Springer: Cham, 2019. [Google Scholar]
- Young G.; Kukura P. Interferometric Scattering Microscopy. Annu. Rev. Phys. Chem. 2019, 70, 301–322. 10.1146/annurev-physchem-050317-021247. [DOI] [PubMed] [Google Scholar]
- Lin Y.-H.; Chang W.-L.; Hsieh C.-L. Shot-Noise Limited Localization of Single 20 Nm Gold Particles with Nanometer Spatial Precision within Microseconds. Opt. Express 2014, 22, 9159–9170. 10.1364/OE.22.009159. [DOI] [PubMed] [Google Scholar]
- Spindler S.; Ehrig J.; König K.; Nowak T.; Piliarik M.; Stein H. E.; Taylor R. W.; Garanger E.; Lecommandoux S.; Alves I. D.; et al. Visualization of Lipids and Proteins at High Spatial and Temporal Resolution via Interferometric Scattering (ISCAT) Microscopy. J. Phys. D: Appl. Phys. 2016, 49, 274002. 10.1088/0022-3727/49/27/274002. [DOI] [Google Scholar]
- Liao Y. H.; Lin C. H.; Cheng C. Y.; Wong W. C.; Juo J. Y.; Hsieh C. L. Monovalent and Oriented Labeling of Gold Nanoprobes for the High-Resolution Tracking of a Single-Membrane Molecule. ACS Nano 2019, 13, 10918–10928. 10.1021/acsnano.9b01176. [DOI] [PubMed] [Google Scholar]
- Spindler S.; Sibold J.; Gholami Mahmoodabadi R.; Steinem C.; Sandoghdar V. High-Speed Microscopy of Diffusion in Pore-Spanning Lipid Membranes. Nano Lett. 2018, 18, 5262–5271. 10.1021/acs.nanolett.8b02240. [DOI] [PubMed] [Google Scholar]
- Andrecka J.; Takagi Y.; Mickolajczyk K. J. J.; Lippert L. G. G.; Sellers J. R. R.; Hancock W. O. O.; Goldman Y. E. E.; Kukura P. Interferometric Scattering Microscopy for the Study of Molecular Motors. Methods Enzymol. 2016, 581, 517–539. 10.1016/bs.mie.2016.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andrecka J.; Ortega Arroyo J.; Takagi Y.; de Wit G.; Fineberg A.; MacKinnon L.; Young G.; Sellers J. R.; Kukura P. Structural Dynamics of Myosin 5 during Processive Motion Revealed by Interferometric Scattering Microscopy. eLife 2015, 4, e05413. 10.7554/eLife.05413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mickolajczyk K. J.; Geyer E. A.; Kim T.; Rice L. M.; Hancock W. O. Direct Observation of Individual Tubulin Dimers Binding to Growing Microtubules. Proc. Natl. Acad. Sci. U. S. A. 2019, 116, 7314–7322. 10.1073/pnas.1815823116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor R. W.; Mahmoodabadi R. G.; Rauschenberger V.; Giessl A.; Schambony A.; Sandoghdar V. Interferometric Scattering Microscopy Reveals Microsecond Nanoscopic Protein Motion on a Live Cell Membrane. Nat. Photonics 2019, 13, 480–487. 10.1038/s41566-019-0414-6. [DOI] [Google Scholar]
- de Wit G.; Albrecht D.; Ewers H.; Kukura P. Revealing Compartmentalized Diffusion in Living Cells with Interferometric Scattering Microscopy. Biophys. J. 2018, 114, 2945–2950. 10.1016/j.bpj.2018.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor R. W.; Holler C.; Mahmoodabadi R. G.; Küppers M.; Dastjerdi H. M.; Zaburdaev V.; Schambony A.; Sandoghdar V. High-Precision Protein-Tracking With Interferometric Scattering Microscopy. Front. Cell Dev. Biol. 2020, 8, 590158. 10.3389/fcell.2020.590158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan M.; Mojarad N.; Kukura P.; Sandoghdar V. Geometry-Induced Electrostatic Trapping of Nanometric Objects in a Fluid. Nature 2010, 467, 692–695. 10.1038/nature09404. [DOI] [PubMed] [Google Scholar]
- Mahmoodabadi R. G.; Taylor R. W.; Kaller M.; Spindler S.; Sandoghdar V. The Point Spread Function in Interferometric Scattering Microscopy (ISCAT). I. Aberrations in Defocusing and Axial Localization. Opt. Express 2020, 28, 25969–25988. 10.1364/OE.401374. [DOI] [PubMed] [Google Scholar]
- Lee I.-B.; Moon H.-M.; Park J.-S.; Zambochova K.; Hong S.-C.; Cho M. Three-Dimensional Interferometric Scattering Microscopy via Remote Focusing Technique. Opt. Lett. 2020, 45, 2628–2631. 10.1364/OL.386172. [DOI] [PubMed] [Google Scholar]
- Cohen A. E.; Moerner W. E. Method for Trapping and Manipulating Nanoscale Objects in Solution. Appl. Phys. Lett. 2005, 86, 093109. 10.1063/1.1872220. [DOI] [Google Scholar]
- Wang Q.; Goldsmith R. H.; Jiang Y.; Bockenhauer S. D.; Moerner W. E. Probing Single Biomolecules in Solution Using the Anti-Brownian Electrokinetic (ABEL) Trap. Acc. Chem. Res. 2012, 45, 1955–1964. 10.1021/ar200304t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Squires A. H.; Lavania A. A.; Dahlberg P. D.; Moerner W. E. Interferometric Scattering Enables Fluorescence-Free Electrokinetic Trapping of Single Nanoparticles in Free Solution. Nano Lett. 2019, 19, 4112–4117. 10.1021/acs.nanolett.9b01514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mojarad N.; Sandoghdar V.; Krishnan M. Measuring Three-Dimensional Interaction Potentials Using Optical Interference. Opt. Express 2013, 21, 9377–9389. 10.1364/OE.21.009377. [DOI] [PubMed] [Google Scholar]
- Tae Kim J.; Spindler S.; Sandoghdar V. Scanning-Aperture Trapping and Manipulation of Single Charged Nanoparticles. Nat. Commun. 2014, 5, 3380. 10.1038/ncomms4380. [DOI] [PubMed] [Google Scholar]
- Tuna Y.; Kim J. T.; Liu H. W.; Sandoghdar V. Levitated Plasmonic Nanoantennas in an Aqueous Environment. ACS Nano 2017, 11, 7674–7678. 10.1021/acsnano.7b03310. [DOI] [PubMed] [Google Scholar]
- Eberle P.; Höller C.; Müller P.; Suomalainen M.; Greber U. F.; Eghlidi H.; Poulikakos D. Single Entity Resolution Valving of Nanoscopic Species in Liquids. Nat. Nanotechnol. 2018, 13, 578–582. 10.1038/s41565-018-0150-y. [DOI] [PubMed] [Google Scholar]
- Skaug M. J.; Schwemmer C.; Fringes S.; Rawlings C. D.; Knoll A. W. Nanofluidic Rocking Brownian Motors. Science 2018, 359, 1505–1508. 10.1126/science.aal3271. [DOI] [PubMed] [Google Scholar]
- Goldfain A. M.; Garmann R. F.; Jin Y.; Lahini Y.; Manoharan V. N. Dynamic Measurements of the Position, Orientation, and DNA Content of Individual Unlabeled Bacteriophages. J. Phys. Chem. B 2016, 120, 6130–6138. 10.1021/acs.jpcb.6b02153. [DOI] [PubMed] [Google Scholar]
- Garmann R. F.; Goldfain A. M.; Manoharan V. N. Measurements of the Self-Assembly Kinetics of Individual Viral Capsids around Their RNA Genome. Proc. Natl. Acad. Sci. U. S. A. 2019, 116, 22485–22490. 10.1073/pnas.1909223116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andrecka J.; Ortega Arroyo J.; Lewis K.; Cross R. A.; Kukura P. Label-Free Imaging of Microtubules with Sub-Nm Precision Using Interferometric Scattering Microscopy. Biophys. J. 2016, 110, 214–217. 10.1016/j.bpj.2015.10.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee I.-B. B.; Moon H.-M. M.; Joo J.-H. H.; Kim K.-H. H.; Hong S.-C. C.; Cho M. Interferometric Scattering Microscopy with Polarization-Selective Dual Detection Scheme: Capturing the Orientational Information of Anisotropic Nanometric Objects. ACS Photonics 2018, 5, 797–804. 10.1021/acsphotonics.7b00890. [DOI] [Google Scholar]
- Vala M.; Bujak Ł.; García Marín A.; Holanová K.; Henrichs V.; Braun M.; Lánský Z.; Piliarik M. Nanoscopic Structural Fluctuations of Disassembling Microtubules Revealed by Label Free Super Resolution Microscopy. Small Methods 2021, 5, 2000985. 10.1002/smtd.202000985. [DOI] [PubMed] [Google Scholar]
- Talà L.; Fineberg A.; Kukura P.; Persat A. Pseudomonas Aeruginosa Orchestrates Twitching Motility by Sequential Control of Type IV Pili Movements. Nat. Microbiol. 2019, 4, 774–780. 10.1038/s41564-019-0378-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Wit G.; Danial J. S. H.; Kukura P.; Wallace M. I. Dynamic Label-Free Imaging of Lipid Nanodomains. Proc. Natl. Acad. Sci. U. S. A. 2015, 112, 12299–12303. 10.1073/pnas.1508483112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andrecka J.; Spillane K. M.; Ortega-Arroyo J.; Kukura P. Direct Observation and Control of Supported Lipid Bilayer Formation with Interferometric Scattering Microscopy. ACS Nano 2013, 7, 10662–10670. 10.1021/nn403367c. [DOI] [PubMed] [Google Scholar]
- Ortega-Arroyo J.; Bissette A. J.; Kukura P.; Fletcher S. P. Visualization of the Spontaneous Emergence of a Complex, Dynamic, and Autocatalytic System. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, 11122–11126. 10.1073/pnas.1602363113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebedeva M. A.; Palmieri E.; Kukura P.; Fletcher S. P. Emergence and Rearrangement of Dynamic Supramolecular Aggregates Visualized by Interferometric Scattering Microscopy. ACS Nano 2020, 14, 11160–11168. 10.1021/acsnano.0c02414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engwerda A. H. J.; Southworth J.; Lebedeva M. A.; Scanes R. J. H.; Kukura P.; Fletcher S. P. Coupled Metabolic Cycles Allow Out-of-Equilibrium Autopoietic Vesicle Replication. Angew. Chem. 2020, 132, 20541–29546. 10.1002/ange.202007302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stetefeld J.; McKenna S. A.; Patel T. R. Dynamic Light Scattering: A Practical Guide and Applications in Biomedical Sciences. Biophys. Rev. 2016, 8, 409–427. 10.1007/s12551-016-0218-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falke S.; Betzel C.. Dynamic Light Scattering (DLS). In Radiation in Bioanalysis; Nature Publishing Group, 2019; Vol. 8, pp 173–193. 10.1007/978-3-030-28247-9_6. [DOI] [Google Scholar]
- McDonald M. P.; Gemeinhardt A.; König K.; Piliarik M.; Schaffer S.; Völkl S.; Aigner M.; Mackensen A.; Sandoghdar V. Visualizing Single-Cell Secretion Dynamics with Single-Protein Sensitivity. Nano Lett. 2018, 18, 513–519. 10.1021/acs.nanolett.7b04494. [DOI] [PubMed] [Google Scholar]
- Gemeinhardt A.; McDonald M. P.; König K.; Aigner M.; Mackensen A.; Sandoghdar V. Label-Free Imaging of Single Proteins Secreted from Living Cells via Iscat Microscopy. J. Visualized Exp. 2018, 2018, 58486. 10.3791/58486. [DOI] [PubMed] [Google Scholar]
- McMeekin T. L.; Wilensky M.; Groves M. L. Refractive Indices of Proteins in Relation to Amino Acid Composition and Specific Volume. Biochem. Biophys. Res. Commun. 1962, 7, 151–156. 10.1016/0006-291X(62)90165-1. [DOI] [Google Scholar]
- Harpaz Y.; Gerstein M.; Chothia C. Volume Changes on Protein Folding. Structure 1994, 2, 641–649. 10.1016/S0969-2126(00)00065-4. [DOI] [PubMed] [Google Scholar]
- Marklund E. G.; Benesch J. L. Weighing-up Protein Dynamics: The Combination of Native Mass Spectrometry and Molecular Dynamics Simulations. Curr. Opin. Struct. Biol. 2019, 2019, 50–58. 10.1016/j.sbi.2018.12.011. [DOI] [PubMed] [Google Scholar]
- Boeri Erba E.; Signor L.; Petosa C. Exploring the Structure and Dynamics of Macromolecular Complexes by Native Mass Spectrometry. J. Proteomics 2020, 222, 103799. 10.1016/j.jprot.2020.103799. [DOI] [PubMed] [Google Scholar]
- Robert H. M. L.; Holanová K.; Bujak Ł.; Vala M.; Henrichs V.; Lánský Z.; Piliarik M. Fast Photothermal Spatial Light Modulation for Quantitative Phase Imaging at the Nanoscale. Nat. Commun. 2021, 12, 2921. 10.1038/s41467-021-23252-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hajdusits B.; Suskiewicz M. J.; Hundt N.; Meinhart A.; Kurzbauer R.; Leodolter J.; Kukura P.; Clausen T. McsB Forms a Gated Kinase Chamber to Mark Aberrant Bacterial Proteins for Degradation. eLife 2021, 10, e63505. 10.7554/eLife.63505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhatt V.; Shi K.; Salamango D. J.; Moeller N. H.; Pandey K. K.; Bera S.; Bohl T. E.; Kurniawan F.; Orellana K.; Zhang W.; et al. Structural Basis of Host Protein Hijacking in Human T-Cell Leukemia Virus Integration. Nat. Commun. 2020, 11, 3121. 10.1038/s41467-020-16963-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- England R. M.; Moss J. I.; Gunnarsson A.; Parker J. S.; Ashford M. B. Synthesis and Characterization of Dendrimer-Based Polysarcosine Star Polymers: Well-Defined, Versatile Platforms Designed for Drug-Delivery Applications. Biomacromolecules 2020, 21, 3332–3341. 10.1021/acs.biomac.0c00768. [DOI] [PubMed] [Google Scholar]
- Fineberg A.; Surrey T.; Kukura P. Quantifying the Monomer–Dimer Equilibrium of Tubulin with Mass Photometry. J. Mol. Biol. 2020, 432, 6168–6172. 10.1016/j.jmb.2020.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye X.; Vogt M. S.; van der Does C.; Bildl W.; Schulte U.; Essen L.-O.; Albers S.-V. The Phosphatase PP2A Interacts With ArnA and ArnB to Regulate the Oligomeric State and the Stability of the ArnA/B Complex. Front. Microbiol. 2020, 11, 1849. 10.3389/fmicb.2020.01849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lonhienne T.; Low Y. S.; Garcia M. D.; Croll T.; Gao Y.; Wang Q.; Brillault L.; Williams C. M.; Fraser J. A.; McGeary R. P.; et al. Structures of Fungal and Plant Acetohydroxyacid Synthases. Nature 2020, 586, 317–321. 10.1038/s41586-020-2514-3. [DOI] [PubMed] [Google Scholar]
- Li Y.; Struwe W. B.; Kukura P. Single Molecule Mass Photometry of Nucleic Acids. Nucleic Acids Res. 2020, 48, e97. 10.1093/nar/gkaa632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Satapathy S.; Dabbs R. A.; Wilson M. R. Rapid High-Yield Expression and Purification of Fully Post-Translationally Modified Recombinant Clusterin and Mutants. Sci. Rep. 2020, 10, 14243. 10.1038/s41598-020-70990-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reikine S.; Mclaughlin S. H.; Modis Y. Equilibrium Binding Model for CpG DNA-Dependent Dimerization of Toll-like Receptor 9 Ectodomain. Biochemistry 2020, 59, 4155–4162. 10.1021/acs.biochem.0c00466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fouët G.; Bally I.; Signor L.; Häußermann K.; Thielens N. M.; Rossi V.; Gaboriaud C. Headless C1q: A New Molecular Tool to Decipher Its Collagen-like Functions. FEBS J. 2021, 288, 2030–2041. 10.1111/febs.15543. [DOI] [PubMed] [Google Scholar]
- Dersch S.; Reimold C.; Stoll J.; Breddermann H.; Heimerl T.; Defeu Soufo H. J.; Graumann P. L. Polymerization of Bacillus Subtilis MreB on a Lipid Membrane Reveals Lateral Co-Polymerization of MreB Paralogs and Strong Effects of Cations on Filament Formation. BMC Mol. Cell Biol. 2020, 21, 76. 10.1186/s12860-020-00319-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malay A. D.; Miyazaki N.; Biela A.; Chakraborti S.; Majsterkiewicz K.; Stupka I.; Kaplan C. S.; Kowalczyk A.; Piette B. M. A. G.; Hochberg G. K. A.; et al. An Ultra-Stable Gold-Coordinated Protein Cage Displaying Reversible Assembly. Nature 2019, 569, 438–442. 10.1038/s41586-019-1185-4. [DOI] [PubMed] [Google Scholar]
- Tjondro H. C.; Ugonotti J.; Kawahara R.; Chatterjee S.; Chen S.; Soltermann F.; Hinneburg H.; Parker B. L.; Venkatakrishnan V.; Loke I.; et al. Hyper-Truncated Asn355- And Asn391-Glycans Modulate the Activity of Neutrophil Granule Myeloperoxidase. J. Biol. Chem. 2021, 296, 100144. 10.1074/jbc.RA120.016342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marcaida M. J.; Kauzlaric A.; Duperrex A.; Sülzle J.; Moncrieffe M. C.; Adebajo D.; Manley S.; Trono D.; Dal Peraro M. The Human RNA Helicase DDX21 Presents a Dimerization Interface Necessary for Helicase Activity. iScience 2020, 23, 101811. 10.1016/j.isci.2020.101811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takada H.; Roghanian M.; Caballero-Montes J.; Van Nerom K.; Jimmy S.; Kudrin P.; Trebini F.; Murayama R.; Akanuma G.; Garcia-Pino A.; et al. Ribosome Association Primes the Stringent Factor Rel for TRNA-Dependent Locking in the A-Site and Activation of (p)PpGpp Synthesis. Nucleic Acids Res. 2021, 49, 444–457. 10.1093/nar/gkaa1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olerinyova A.; Sonn-Segev A.; Gault J.; Eichmann C.; Schimpf J.; Kopf A. H.; Rudden L. S. P.; Ashkinadze D.; Bomba R.; Frey L.; et al. Mass Photometry of Membrane Proteins. Chem. 2021, 7, 224–236. 10.1016/j.chempr.2020.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilk P.; Kuśka K.; Wa̧tor E.; Małecki P. H.; Woś K.; Tokarz P.; Dubin G.; Grudnik P. Structural Characterization of Glycerol Kinase from the Thermophilic Fungus Chaetomium Thermophilum. Int. J. Mol. Sci. 2020, 21, 9570. 10.3390/ijms21249570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gunnarsson A.; Stubbs C. J.; Rawlins P. B.; Taylor-Newman E.; Lee W. C.; Geschwindner S.; Hytönen V.; Holdgate G.; Jha R.; Dahl G. Regenerable Biosensors for Small-Molecule Kinetic Characterization Using SPR. SLAS Discovery 2021, 26, 730–739. 10.1177/2472555220975358. [DOI] [PubMed] [Google Scholar]
- Wu D.; Piszczek G. Rapid Determination of Antibody-Antigen Affinity by Mass Photometry. J. Visualized Exp. 2021, 168, e61784 10.3791/61784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thabault L.; Liberelle M.; Koruza K.; Yildiz E.; Joudiou N.; Messens J.; Brisson L.; Wouters J.; Sonveaux P.; Frédérick R. Discovery of a Novel Lactate Dehydrogenase Tetramerization Domain Using Epitope Mapping and Peptides. J. Biol. Chem. 2021, 296, 100422. 10.1016/j.jbc.2021.100422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foley E. D. B.; Kushwah M. S.; Young G.; Kukura P.. Label-Free Tracking and Mass Measurement of Single Proteins on Lipid Bilayers. bioRxiv 2021, 2021.04.08.438951. 10.1101/2021.04.08.438951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heermann T.; Steiert F.; Ramm B.; Hundt N.; Schwille P.. Mass-Sensitive Particle Tracking (MSPT) to Elucidate the Membrane-Associated MinDE Reaction Cycle. bioRxiv 2021, 2021.04.08.438961. 10.1101/2021.04.08.438961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Häußermann K.; Young G.; Kukura P.; Dietz H. Dissecting FOXP2 Oligomerization and DNA Binding. Angew. Chem., Int. Ed. 2019, 58, 7662–7667. 10.1002/anie.201901734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hickman A. B.; Kailasan S.; Genzor P.; Haase A. D.; Dyda F. Casposase Structure and the Mechanistic Link between DNA Transposition and Spacer Acquisition by CRISPR-Cas. eLife 2020, 9, e50004. 10.7554/eLife.50004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonn-Segev A.; Belacic K.; Bodrug T.; Young G.; VanderLinden R. T.; Schulman B. A.; Schimpf J.; Friedrich T.; Dip P. V.; Schwartz T. U.; et al. Quantifying the Heterogeneity of Macromolecular Machines by Mass Photometry. Nat. Commun. 2020, 11, 1772. 10.1038/s41467-020-15642-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu D.; Piszczek G. Measuring the Affinity of Protein-Protein Interactions on a Single-Molecule Level by Mass Photometry. Anal. Biochem. 2020, 592, 113575. 10.1016/j.ab.2020.113575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soltermann F.; Foley E. D. B.; Pagnoni V.; Galpin M.; Benesch J. L. P.; Kukura P.; Struwe W. B. Quantifying Protein–Protein Interactions by Molecular Counting with Mass Photometry. Angew. Chem., Int. Ed. 2020, 59, 10774–10779. 10.1002/anie.202001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres-Ocampo A. P.; Özden C.; Hommer A.; Gardella A.; Lapinskas E.; Samkutty A.; Esposito E.; Garman S. C.; Stratton M. M. Characterization of CaMKIIα Holoenzyme Stability. Protein Sci. 2020, 29, 1524–1534. 10.1002/pro.3869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tamara S.; Franc V.; Heck A. J. R. A Wealth of Genotype-Specific Proteoforms Fine-Tunes Hemoglobin Scavenging by Haptoglobin. Proc. Natl. Acad. Sci. U. S. A. 2020, 117, 15554–15564. 10.1073/pnas.2002483117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Avci O.; Ünlü N.; Özkumur A.; Ünlü M. Interferometric Reflectance Imaging Sensor (IRIS)—A Platform Technology for Multiplexed Diagnostics and Digital Detection. Sensors 2015, 15, 17649–17665. 10.3390/s150717649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Özkumur E.; Needham J. W.; Bergstein D. A.; Gonzalez R.; Cabodi M.; Gershoni J. M.; Goldberg B. B.; Ünlü M. S. Label-Free and Dynamic Detection of Biomolecular Interactions for High-Throughput Microarray Applications. Proc. Natl. Acad. Sci. U. S. A. 2008, 105, 7988–7992. 10.1073/pnas.0711421105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daaboul G. G.; Yurt A.; Zhang X.; Hwang G. M.; Goldberg B. B.; Ünlü M. S. High-Throughput Detection and Sizing of Individual Low-Index Nanoparticles and Viruses for Pathogen Identification. Nano Lett. 2010, 10, 4727–4731. 10.1021/nl103210p. [DOI] [PubMed] [Google Scholar]
- Sevenler D.; Avci O.; Ünlü M. S. Quantitative Interferometric Reflectance Imaging for the Detection and Measurement of Biological Nanoparticles. Biomed. Opt. Express 2017, 8, 2976–2989. 10.1364/BOE.8.002976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sevenler D.; Daaboul G. G.; Ekiz Kanik F.; Ünlü N. L.; Ünlü M. S. Digital Microarrays: Single-Molecule Readout with Interferometric Detection of Plasmonic Nanorod Labels. ACS Nano 2018, 12, 5880–5887. 10.1021/acsnano.8b02036. [DOI] [PubMed] [Google Scholar]
- Daaboul G. G.; Lopez C. A.; Chinnala J.; Goldberg B. B.; Connor J. H.; Ünlü M. S. Digital Sensing and Sizing of Vesicular Stomatitis Virus Pseudotypes in Complex Media: A Model for Ebola and Marburg Detection. ACS Nano 2014, 8, 6047–6055. 10.1021/nn501312q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daaboul G. G.; Freedman D. S.; Scherr S. M.; Carter E.; Rosca A.; Bernstein D.; Mire C. E.; Agans K. N.; Hoenen T.; Geisbert T. W.; et al. Enhanced Light Microscopy Visualization of Virus Particles from Zika Virus to Filamentous Ebolaviruses. PLoS One 2017, 12, e0179728 10.1371/journal.pone.0179728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Daaboul G. G.; Gagni P.; Benussi L.; Bettotti P.; Ciani M.; Cretich M.; Freedman D. S.; Ghidoni R.; Ozkumur A. Y.; Piotto C.; et al. Digital Detection of Exosomes by Interferometric Imaging. Sci. Rep. 2016, 6, 37246. 10.1038/srep37246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scherr S. M.; Daaboul G. G.; Trueb J.; Sevenler D.; Fawcett H.; Goldberg B.; Connor J. H.; Ünlü M. S. Real-Time Capture and Visualization of Individual Viruses in Complex Media. ACS Nano 2016, 10, 2827–2833. 10.1021/acsnano.5b07948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scherr S. M.; Freedman D. S.; Agans K. N.; Rosca A.; Carter E.; Kuroda M.; Fawcett H. E.; Mire C. E.; Geisbert T. W.; Ünlü M. S.; et al. Disposable Cartridge Platform for Rapid Detection of Viral Hemorrhagic Fever Viruses. Lab Chip 2017, 17, 917–925. 10.1039/C6LC01528J. [DOI] [PubMed] [Google Scholar]
- Seymour E.; Daaboul G. G.; Zhang X.; Scherr S. M.; Ünlü N. L.; Connor J. H.; Ünlü M. S. DNA-Directed Antibody Immobilization for Enhanced Detection of Single Viral Pathogens. Anal. Chem. 2015, 87, 10505–10512. 10.1021/acs.analchem.5b02702. [DOI] [PubMed] [Google Scholar]
- Seymour E.; Ünlü N. L.; Carter E. P.; Connor J. H.; Ünlü M. S. Configurable Digital Virus Counter on Robust Universal DNA Chips. ACS Sensors 2021, 6, 229–237. 10.1021/acssensors.0c02203. [DOI] [PubMed] [Google Scholar]
- Trueb J. T.; Avci O.; Sevenler D.; Connor J. H.; Ünlü M. S. Robust Visualization and Discrimination of Nanoparticles by Interferometric Imaging. IEEE J. Sel. Top. Quantum Electron. 2017, 23, 394–403. 10.1109/JSTQE.2016.2639824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aygun U.; Urey H.; Yalcin Ozkumur A. Label-Free Detection of Nanoparticles Using Depth Scanning Correlation Interferometric Microscopy. Sci. Rep. 2019, 9, 9012. 10.1038/s41598-019-45439-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yurdakul C.; Ünlü M. S. Computational Nanosensing from Defocus in Single Particle Interferometric Reflectance Microscopy. Opt. Lett. 2020, 45, 6546–6549. 10.1364/OL.409458. [DOI] [PubMed] [Google Scholar]
- Tian L.; Waller L. Quantitative Differential Phase Contrast Imaging in an LED Array Microscope. Opt. Express 2015, 23, 11394–11403. 10.1364/OE.23.011394. [DOI] [PubMed] [Google Scholar]
- Tian L.; Li X.; Ramchandran K.; Waller L. Multiplexed Coded Illumination for Fourier Ptychography with an LED Array Microscope. Biomed. Opt. Express 2014, 5, 2376–2389. 10.1364/BOE.5.002376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yurdakul C.; Avci O.; Matlock A.; Devaux A. J.; Quintero M. V.; Ozbay E.; Davey R. A.; Connor J. H.; Karl W. C.; Tian L. High-Throughput, High-Resolution Interferometric Light Microscopy of Biological Nanoparticles. ACS Nano 2020, 14, 2002–2013. 10.1021/acsnano.9b08512. [DOI] [PubMed] [Google Scholar]
- Monroe M. R.; Daaboul G. G.; Tuysuzoglu A.; Lopez C. A.; Little F. F.; Ünlü M. S. Single Nanoparticle Detection for Multiplexed Protein Diagnostics with Attomolar Sensitivity in Serum and Unprocessed Whole Blood. Anal. Chem. 2013, 85, 3698–3706. 10.1021/ac4000514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sevenler D.; Trueb J.; Ünlu M. S. Beating the Reaction Limits of Biosensor Sensitivity with Dynamic Tracking of Single Binding Events. Proc. Natl. Acad. Sci. U. S. A. 2019, 116, 4129–4134. 10.1073/pnas.1815329116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang Y.-F.; Zhuo G.-Y.; Chou C.-Y.; Lin C.-H.; Chang W.; Hsieh C.-L. Coherent Brightfield Microscopy Provides the Spatiotemporal Resolution To Study Early Stage Viral Infection in Live Cells. ACS Nano 2017, 11, 2575–2585. 10.1021/acsnano.6b05601. [DOI] [PubMed] [Google Scholar]
- Cheng C.-Y.; Liao Y.-H.; Hsieh C.-L. High-Speed Imaging and Tracking of Very Small Single Nanoparticles by Contrast Enhanced Microscopy. Nanoscale 2019, 11, 568–577. 10.1039/C8NR06789A. [DOI] [PubMed] [Google Scholar]
- Huang Y. F.; Zhuo G. Y.; Chou C. Y.; Lin C. H.; Hsieh C. L. Label-Free, Ultrahigh-Speed, 3D Observation of Bidirectional and Correlated Intracellular Cargo Transport by Coherent Brightfield Microscopy. Nanoscale 2017, 9, 6567–6574. 10.1039/C7NR00604G. [DOI] [PubMed] [Google Scholar]
- Liedberg B.; Nylander C.; Lunström I. Surface Plasmon Resonance for Gas Detection and Biosensing. Sens. Actuators 1983, 4, 299–304. 10.1016/0250-6874(83)85036-7. [DOI] [Google Scholar]
- Fischer B.; Heyn S. P.; Egger M.; Gaub H. E. Antigen Binding to a Pattern of Lipid-Anchored Antibody Binding Sites Measured by Surface Plasmon Microscopy. Langmuir 1993, 9, 136–140. 10.1021/la00025a031. [DOI] [Google Scholar]
- Berger C. E. H.; Beumer T. A. M.; Kooyman R. P. H.; Greve J. Surface Plasmon Resonance Multisensing. Anal. Chem. 1998, 70, 703–706. 10.1021/ac970929a. [DOI] [Google Scholar]
- Ladd J.; Taylor A. D.; Piliarik M.; Homola J.; Jiang S. Label-Free Detection of Cancer Biomarker Candidates Using Surface Plasmon Resonance Imaging. Anal. Bioanal. Chem. 2009, 393, 1157–1163. 10.1007/s00216-008-2448-3. [DOI] [PubMed] [Google Scholar]
- Liu W.; Chen Y.; Yan M. Surface Plasmon Resonance Imaging of Limited Glycoprotein Samples. Analyst 2008, 133, 1268–1273. 10.1039/b804235g. [DOI] [PubMed] [Google Scholar]
- Rebe Raz S.; Bremer M. G. E. G.; Giesbers M.; Norde W. Development of a Biosensor Microarray towards Food Screening, Using Imaging Surface Plasmon Resonance. Biosens. Bioelectron. 2008, 24, 552–557. 10.1016/j.bios.2008.05.010. [DOI] [PubMed] [Google Scholar]
- Wong C. L.; Ho H. P.; Suen Y. K.; Kong S. K.; Chen Q. L.; Yuan W.; Wu S. Y. Real-Time Protein Biosensor Arrays Based on Surface Plasmon Resonance Differential Phase Imaging. Biosens. Bioelectron. 2008, 24, 606–612. 10.1016/j.bios.2008.06.013. [DOI] [PubMed] [Google Scholar]
- Yuk J. S.; Kim H. S.; Jung J. W.; Jung S. H.; Lee S. J.; Kim W. J.; Han J. A.; Kim Y. M.; Ha K. S. Analysis of Protein Interactions on Protein Arrays by a Novel Spectral Surface Plasmon Resonance Imaging. Biosens. Bioelectron. 2006, 21, 1521–1528. 10.1016/j.bios.2005.07.009. [DOI] [PubMed] [Google Scholar]
- Wegner G. J.; Lee H. J.; Marriott G.; Corn R. M. Fabrication of Histidine-Tagged Fusion Protein Arrays for Surface Plasmon Resonance Imaging Studies of Protein - Protein and Protein - DNA Interactions. Anal. Chem. 2003, 75, 4740–4746. 10.1021/ac0344438. [DOI] [PubMed] [Google Scholar]
- Jordan C. E.; Frutos A. G.; Thiel A. J.; Corn R. M. Surface Plasmon Resonance Imaging Measurements of DNA Hybridization Adsorption and Streptavidin/DNA Multilayer Formation at Chemically Modified Gold Surfaces. Anal. Chem. 1997, 69, 4939–4947. 10.1021/ac9709763. [DOI] [PubMed] [Google Scholar]
- Nelson B. P.; Grimsrud T. E.; Liles M. R.; Goodman R. M.; Corn R. M. Surface Plasmon Resonance Imaging Measurements of DNA and RNA Hybridization Adsorption onto DNA Microarrays. Anal. Chem. 2001, 73, 1–7. 10.1021/ac0010431. [DOI] [PubMed] [Google Scholar]
- Mannelli I.; Courtois V.; Lecaruyer P.; Roger G.; Millot M. C.; Goossens M.; Canva M. Surface Plasmon Resonance Imaging (SPRI) System and Real-Time Monitoring of DNA Biochip for Human Genetic Mutation Diagnosis of DNA Amplified Samples. Sens. Actuators, B 2006, 119, 583–591. 10.1016/j.snb.2006.01.023. [DOI] [Google Scholar]
- Hayashi G.; Hagihara M.; Nakatani K. Genotyping by Allele-Specific l-DNA-Tagged PCR. J. Biotechnol. 2008, 135, 157–160. 10.1016/j.jbiotec.2008.03.011. [DOI] [PubMed] [Google Scholar]
- Li Y.; Lee H. J.; Corn R. M. Fabrication and Characterization of RNA Aptamer Microarrays for the Study of Protein-Aptamer Interactions with SPR Imaging. Nucleic Acids Res. 2006, 34, 6416–6424. 10.1093/nar/gkl738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson B. P.; Liles M. R.; Frederick K. B.; Corn R. M.; Goodman R. M. Label-Free Detection of 16S Ribosomal RNA Hybridization on Reusable DNA Arrays Using Surface Plasmon Resonance Imaging. Environ. Microbiol. 2002, 4, 735–743. 10.1046/j.1462-2920.2002.00350.x. [DOI] [PubMed] [Google Scholar]
- Nelson B. P.; Frutos A. G.; Brockman J. M.; Corn R. M. Near-Infrared Surface Plasmon Resonance Measurements of Ultrathin Films. 1. Angle Shift and SPR Imaging Experiments. Anal. Chem. 1999, 71, 3928–3934. 10.1021/ac990517x. [DOI] [Google Scholar]
- Brockman J. M.; Frutos A. G.; Corn R. M. A Multistep Chemical Modification Procedure to Create Dna Arrays on Gold Surfaces for the Study of Protein-DNA Interactions with Surface Plasmon Resonance Imaging. J. Am. Chem. Soc. 1999, 121, 8044–8051. 10.1021/ja991608e. [DOI] [Google Scholar]
- Jeong E. J.; Jeong Y. S.; Park K.; Yi S. Y.; Ahn J.; Chung S. J.; Kim M.; Chung B. H. Directed Immobilization of DNA-Binding Proteins on a Cognate DNA-Modified Chip Surface. J. Biotechnol. 2008, 135, 16–21. 10.1016/j.jbiotec.2008.02.019. [DOI] [PubMed] [Google Scholar]
- Homola J.; Yee S. S.; Gauglitz G. Surface Plasmon Resonance Sensors: Review. Sens. Actuators, B 1999, 54, 3–15. 10.1016/S0925-4005(98)00321-9. [DOI] [Google Scholar]
- Homola J. Surface Plasmon Resonance Sensors for Detection of Chemical and Biological Species. Chem. Rev. 2008, 108, 462–493. 10.1021/cr068107d. [DOI] [PubMed] [Google Scholar]
- Hinman S. S.; McKeating K. S.; Cheng Q. Surface Plasmon Resonance: Material and Interface Design for Universal Accessibility. Anal. Chem. 2018, 90, 19–39. 10.1021/acs.analchem.7b04251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo X. Surface Plasmon Resonance Based Biosensor Technique: A Review. Journal of Biophotonics. 2012, 5, 483–501. 10.1002/jbio.201200015. [DOI] [PubMed] [Google Scholar]
- Masson J. F. Surface Plasmon Resonance Clinical Biosensors for Medical Diagnostics. ACS Sensors. 2017, 2, 16–30. 10.1021/acssensors.6b00763. [DOI] [PubMed] [Google Scholar]
- Zeidan E.; Kepley C. L.; Sayes C.; Sandros M. G. Surface Plasmon Resonance: A Label-Free Tool for Cellular Analysis. Nanomedicine 2015, 10, 1833–1846. 10.2217/nnm.15.31. [DOI] [PubMed] [Google Scholar]
- Stahelin R. V. Surface Plasmon Resonance: A Useful Technique for Cell Biologists to Characterize Biomolecular Interactions. Mol. Biol. Cell 2013, 24, 883–886. 10.1091/mbc.e12-10-0713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jung L. S.; Campbell C. T.; Chinowsky T. M.; Mar M. N.; Yee S. S. Quantitative Interpretation of the Response of Surface Plasmon Resonance Sensors to Adsorbed Films. Langmuir 1998, 14, 5636–5648. 10.1021/la971228b. [DOI] [Google Scholar]
- Yeatman E.; Ash E. A. Surface Plasmon Microscopy. Electron. Lett. 1987, 23, 1091–1092. 10.1049/el:19870762. [DOI] [Google Scholar]
- Rothenhäusler B.; Knoll W. Surface-Plasmon Microscopy. Nature 1988, 332, 615–617. 10.1038/332615a0. [DOI] [Google Scholar]
- Zhang F.; Wang S.; Yin L.; Yang Y.; Guan Y.; Wang W.; Xu H.; Tao N. Quantification of Epidermal Growth Factor Receptor Expression Level and Binding Kinetics on Cell Surfaces by Surface Plasmon Resonance Imaging. Anal. Chem. 2015, 87, 9960–9965. 10.1021/acs.analchem.5b02572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scarano S.; Mascini M.; Turner A. P. F.; Minunni M. Surface Plasmon Resonance Imaging for Affinity-Based Biosensors. Biosens. Bioelectron. 2010, 25, 957–966. 10.1016/j.bios.2009.08.039. [DOI] [PubMed] [Google Scholar]
- Campbell C. T.; Kim G. SPR Microscopy and Its Applications to High-Throughput Analyses of Biomolecular Binding Events and Their Kinetics. Biomaterials 2007, 28, 2380–2392. 10.1016/j.biomaterials.2007.01.047. [DOI] [PubMed] [Google Scholar]
- Fasoli J. B.; Corn R. M. Surface Enzyme Chemistries for Ultrasensitive Microarray Biosensing with SPR Imaging. Langmuir 2015, 31, 9527–9536. 10.1021/la504797z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hinman S. S.; Ruiz C. J.; Drakakaki G.; Wilkop T. E.; Cheng Q. On-Demand Formation of Supported Lipid Membrane Arrays by Trehalose-Assisted Vesicle Delivery for SPR Imaging. ACS Appl. Mater. Interfaces 2015, 7, 17122–17130. 10.1021/acsami.5b03809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manuel G.; Lupták A.; Corn R. M. A Microwell-Printing Fabrication Strategy for the On-Chip Templated Biosynthesis of Protein Microarrays for Surface Plasmon Resonance Imaging. J. Phys. Chem. C 2016, 120, 20984–20990. 10.1021/acs.jpcc.6b03307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang B.; Yu F.; Zare R. N. Surface Plasmon Resonance Imaging Using a High Numerical Aperture Microscope Objective. Anal. Chem. 2007, 79, 2979–2983. 10.1021/ac062284x. [DOI] [PubMed] [Google Scholar]
- Kretschmann E.; Raether H. Radiative Decay of Non Radiative Surface Plasmons Excited by Light. Z. Naturforsch., A: Phys. Sci. 1968, 23, 2135–2136. 10.1515/zna-1968-1247. [DOI] [Google Scholar]
- Orlowski R.; Raether H. The Total Reflection of Light at Smooth and Rough Silver Films and Surface Plasmons. Surf. Sci. 1976, 54, 303–308. 10.1016/0039-6028(76)90227-2. [DOI] [Google Scholar]
- Jiang Y.; Wang W. Point Spread Function of Objective-Based Surface Plasmon Resonance Microscopy. Anal. Chem. 2018, 90, 9650–9656. 10.1021/acs.analchem.8b02800. [DOI] [PubMed] [Google Scholar]
- Yu H.; Shan X.; Wang S.; Chen H.; Tao N. Molecular Scale Origin of Surface Plasmon Resonance Biosensors. Anal. Chem. 2014, 86, 8992–8997. 10.1021/ac501363z. [DOI] [PubMed] [Google Scholar]
- Bozhevolnyi S. I. Localization Phenomena in Elastic Surface-Polariton Scattering Caused by Surface Roughness. Phys. Rev. B: Condens. Matter Mater. Phys. 1996, 54, 8177–8185. 10.1103/PhysRevB.54.8177. [DOI] [PubMed] [Google Scholar]
- Bozhevolnyi S. I.; Coello V. Elastic Scattering of Surface Plasmon Polaritons: Modeling and Experiment. Phys. Rev. B: Condens. Matter Mater. Phys. 1998, 58, 10899–10910. 10.1103/PhysRevB.58.10899. [DOI] [Google Scholar]
- Hecht B.; Bielefeldt H.; Novotny L.; Inouye Y.; Pohl D. W. Local Excitation, Scattering, and Interference of Surface Plasmons. Phys. Rev. Lett. 1996, 77, 1889–1892. 10.1103/PhysRevLett.77.1889. [DOI] [PubMed] [Google Scholar]
- Berini P. Long-Range Surface Plasmon Polaritons. Adv. Opt. Photonics 2009, 1, 484–588. 10.1364/AOP.1.000484. [DOI] [Google Scholar]
- Kelly K. L.; Coronado E.; Zhao L. L.; Schatz G. C. The Optical Properties of Metal Nanoparticles: The Influence of Size, Shape, and Dielectric Environment. J. Phys. Chem. B 2003, 107, 668–677. 10.1021/jp026731y. [DOI] [Google Scholar]
- Becker J.; Trügler A.; Jakab A.; Hohenester U.; Sönnichsen C. The Optimal Aspect Ratio of Gold Nanorods for Plasmonic Bio-Sensing. Plasmonics 2010, 5, 161–167. 10.1007/s11468-010-9130-2. [DOI] [Google Scholar]
- Zijlstra P.; Paulo P. M. R.; Orrit M. Optical Detection of Single Non-Absorbing Molecules Using the Surface Plasmon Resonance of a Gold Nanorod. Nat. Nanotechnol. 2012, 7, 379–382. 10.1038/nnano.2012.51. [DOI] [PubMed] [Google Scholar]
- Ament I.; Prasad J.; Henkel A.; Schmachtel S.; Sönnichsen C. Single Unlabeled Protein Detection on Individual Plasmonic Nanoparticles. Nano Lett. 2012, 12, 1092–1095. 10.1021/nl204496g. [DOI] [PubMed] [Google Scholar]
- Vecchi G.; Giannini V.; Gómez Rivas J. Surface Modes in Plasmonic Crystals Induced by Diffractive Coupling of Nanoantennas. Phys. Rev. B: Condens. Matter Mater. Phys. 2009, 80, 201401. 10.1103/PhysRevB.80.201401. [DOI] [Google Scholar]
- Chen P.; Chung M. T.; McHugh W.; Nidetz R.; Li Y.; Fu J.; Cornell T. T.; Shanley T. P.; Kurabayashi K. Multiplex Serum Cytokine Immunoassay Using Nanoplasmonic Biosensor Microarrays. ACS Nano 2015, 9, 4173–4181. 10.1021/acsnano.5b00396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X.; Soler M.; Özdemir C. I.; Belushkin A.; Yesilköy F.; Altug H. Plasmonic Nanohole Array Biosensor for Label-Free and Real-Time Analysis of Live Cell Secretion. Lab Chip 2017, 17, 2208–2217. 10.1039/C7LC00277G. [DOI] [PubMed] [Google Scholar]
- Guo H.; Guo J. Hybrid Plasmon Photonic Crystal Resonance Grating for Integrated Spectrometer Biosensor. Opt. Lett. 2015, 40, 249–252. 10.1364/OL.40.000249. [DOI] [PubMed] [Google Scholar]
- Seiler S. T.; Rich I. S.; Lindquist N. C. Direct Spectral Imaging of Plasmonic Nanohole Arrays for Real-Time Sensing. Nanotechnology 2016, 27, 184001. 10.1088/0957-4484/27/18/184001. [DOI] [PubMed] [Google Scholar]
- Bockova M.; Slaby J.; Springer T.; Homola J. Advances in Surface Plasmon Resonance Imaging and Microscopy and Their Biological Applications. Annu. Rev. Anal. Chem. 2019, 12, 151–176. 10.1146/annurev-anchem-061318-115106. [DOI] [PubMed] [Google Scholar]
- Zeng Y.; Hu R.; Wang L.; Gu D.; He J.; Wu S. Y.; Ho H. P.; Li X.; Qu J.; Gao B. Z.; et al. Recent Advances in Surface Plasmon Resonance Imaging: Detection Speed, Sensitivity, and Portability. Nanophotonics 2017, 6, 1017–1030. 10.1515/nanoph-2017-0022. [DOI] [Google Scholar]
- Wong C. L.; Olivo M. Surface Plasmon Resonance Imaging Sensors: A Review. Plasmonics 2014, 9, 809–824. 10.1007/s11468-013-9662-3. [DOI] [Google Scholar]
- Wang S.; Shan X.; Patel U.; Huang X.; Lu J.; Li J.; Tao N. Label-Free Imaging, Detection, and Mass Measurement of Single Viruses by Surface Plasmon Resonance. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 16028–16032. 10.1073/pnas.1005264107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Demetriadou A.; Kornyshev A. A. Principles of Nanoparticle Imaging Using Surface Plasmons. New J. Phys. 2015, 17, 013041. 10.1088/1367-2630/17/1/013041. [DOI] [Google Scholar]
- Qian C.; Wu G.; Jiang D.; Zhao X.; Chen H. B.; Yang Y.; Liu X. W. Identification of Nanoparticles via Plasmonic Scattering Interferometry. Angew. Chem., Int. Ed. 2019, 58, 4217–4220. 10.1002/anie.201813567. [DOI] [PubMed] [Google Scholar]
- Yang Y.; Yu H.; Shan X.; Wang W.; Liu X.; Wang S.; Tao N. Label-Free Tracking of Single Organelle Transportation in Cells with Nanometer Precision Using a Plasmonic Imaging Technique. Small 2015, 11, 2878–2884. 10.1002/smll.201403016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu H.; Shan X.; Wang S.; Chen H.; Tao N. Plasmonic Imaging and Detection of Single DNA Molecules. ACS Nano 2014, 8, 3427–3433. 10.1021/nn4062885. [DOI] [PubMed] [Google Scholar]
- Syal K.; Iriya R.; Yang Y.; Yu H.; Wang S.; Haydel S. E.; Chen H. Y.; Tao N. Antimicrobial Susceptibility Test with Plasmonic Imaging and Tracking of Single Bacterial Motions on Nanometer Scale. ACS Nano 2016, 10, 845–852. 10.1021/acsnano.5b05944. [DOI] [PubMed] [Google Scholar]
- Wang W.; Wang S.; Liu Q.; Wu J.; Tao N. Mapping Single-Cell-Substrate Interactions by Surface Plasmon Resonance Microscopy. Langmuir 2012, 28, 13373–13379. 10.1021/la301712h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toma K.; Kano H.; Offenhäusser A. Label-Free Measurement of Cell-Electrode Cleft Gap Distance with High Spatial Resolution Surface Plasmon Microscopy. ACS Nano 2014, 8, 12612–12619. 10.1021/nn505521e. [DOI] [PubMed] [Google Scholar]
- Yang C. T.; Méjard R.; Griesser H. J.; Bagnaninchi P. O.; Thierry B. Cellular Micromotion Monitored by Long-Range Surface Plasmon Resonance with Optical Fluctuation Analysis. Anal. Chem. 2015, 87, 1456–1461. 10.1021/ac5031978. [DOI] [PubMed] [Google Scholar]
- Zhou X.; Yang Y.; Wang S.; Liu X. Surface Plasmon Resonance Microscopy: From Single Molecule Sensing to Single Cell Imaging. Angew. Chem., Int. Ed. 2020, 59, 1776–1785. 10.1002/anie.201908806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foley K. J.; Shan X.; Tao N. J. Surface Impedance Imaging Technique. Anal. Chem. 2008, 80, 5146–5151. 10.1021/ac800361p. [DOI] [PubMed] [Google Scholar]
- Yuan L.; Tao N.; Wang W. Plasmonic Imaging of Electrochemical Impedance. Annu. Rev. Anal. Chem. 2017, 10, 183–200. 10.1146/annurev-anchem-061516-045150. [DOI] [PubMed] [Google Scholar]
- Liang W.; Wang S.; Festa F.; Wiktor P.; Wang W.; Magee M.; Labaer J.; Tao N. Measurement of Small Molecule Binding Kinetics on a Protein Microarray by Plasmonic-Based Electrochemical Impedance Imaging. Anal. Chem. 2014, 86, 9860–9865. 10.1021/ac5024556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Macgriff C.; Wang S.; Wiktor P.; Wang W.; Shan X.; Tao N. Charge-Based Detection of Small Molecules by Plasmonic-Based Electrochemical Impedance Microscopy. Anal. Chem. 2013, 85, 6682–6687. 10.1021/ac400475z. [DOI] [PubMed] [Google Scholar]
- Halpern A. R.; Wood J. B.; Wang Y.; Corn R. M. Single-Nanoparticle Near-Infrared Surface Plasmon Resonance Microscopy for Real-Time Measurements of DNA Hybridization Adsorption. ACS Nano 2014, 8, 1022–1030. 10.1021/nn405868e. [DOI] [PubMed] [Google Scholar]
- Shan X.; Fang Y.; Wang S.; Guan Y.; Chen H. Y.; Tao N. Detection of Charges and Molecules with Self-Assembled Nano-Oscillators. Nano Lett. 2014, 14, 4151–4157. 10.1021/nl501805e. [DOI] [PubMed] [Google Scholar]
- Fang Y.; Chen S.; Wang W.; Shan X.; Tao N. Real-Time Monitoring of Phosphorylation Kinetics with Self-Assembled Nano-Oscillators. Angew. Chem., Int. Ed. 2015, 54, 2538–2542. 10.1002/anie.201411040. [DOI] [PubMed] [Google Scholar]
- Chen Z.; Peng Y.; Cao Y.; Wang H.; Zhang J. R.; Chen H. Y.; Zhu J. J. Light-Driven Nano-Oscillators for Label-Free Single-Molecule Monitoring of MicroRNA. Nano Lett. 2018, 18, 3759–3765. 10.1021/acs.nanolett.8b00993. [DOI] [PubMed] [Google Scholar]
- Ma G.; Wan Z.; Yang Y.; Zhang P.; Wang S.; Tao N. Optical Imaging of Single-Protein Size, Charge, Mobility, and Binding. Nat. Commun. 2020, 11, 4758. 10.1038/s41467-020-18547-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kano H.; Knoll W. A Scanning Microscope Employing Localized Surface-Plasmon-Polaritons as a Sensing Probe. Opt. Commun. 2000, 182, 11–15. 10.1016/S0030-4018(00)00794-X. [DOI] [Google Scholar]
- Watanabe K.; Horiguchi N.; Kano H. Optimized Measurement Probe of the Localized Surface Plasmon Microscope by Using Radially Polarized Illumination. Appl. Opt. 2007, 46, 4985–4990. 10.1364/AO.46.004985. [DOI] [PubMed] [Google Scholar]
- Watanabe K.; Ryosuke M.; Terakado G.; Okazaki T.; Morigaki K.; Kano H. High Resolution Imaging of Patterned Model Biological Membranes by Localized Surface Plasmon Microscopy. Appl. Opt. 2010, 49, 887–891. 10.1364/AO.49.000887. [DOI] [PubMed] [Google Scholar]
- Kuai Y.; Chen J.; Tang X.; Xiang Y.; Lu F.; Kuang C.; Xu L.; Shen W.; Cheng J.; Gui H. Label-Free Surface-Sensitive Photonic Microscopy with High Spatial Resolution Using Azimuthal Rotation Illumination. Sci. Adv. 2019, 5, eaav5335. 10.1126/sciadv.aav5335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banville F. A.; Söllradl T.; Zermatten P.-J.; Grandbois M.; Charette P. G. Improved Resolution in SPR and MCWG Microscopy by Combining Images Acquired with Distinct Mode Propagation Directions. Opt. Lett. 2015, 40, 1165-1168. 10.1364/OL.40.001165. [DOI] [PubMed] [Google Scholar]
- Son T.; Lee C.; Seo J.; Choi I.-H.; Kim D. Surface Plasmon Microscopy by Spatial Light Switching for Label-Free Imaging with Enhanced Resolution. Opt. Lett. 2018, 43, 959–962. 10.1364/OL.43.000959. [DOI] [PubMed] [Google Scholar]
- Tan H. M.; Pechprasarn S.; Zhang J.; Pitter M. C.; Somekh M. G. High Resolution Quantitative Angle-Scanning Widefield Surface Plasmon Microscopy. Sci. Rep. 2016, 6, 20195. 10.1038/srep20195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang W. T.; Chung E.; Kim Y.-H.; So P. T. C.; Sheppard C. J. R. Investigation of the Point Spread Function of Surface Plasmon-Coupled Emission Microscopy. Opt. Express 2007, 15, 4634–4646. 10.1364/OE.15.004634. [DOI] [PubMed] [Google Scholar]
- Tang W. T.; Chung E.; Kim Y.-H.; So P. T.; Sheppard C. J. Surface-Plasmon-Coupled Emission Microscopy with a Spiral Phase Plate. Opt. Lett. 2010, 35, 517–519. 10.1364/OL.35.000517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y.; Zhang D.; Han L.; Rui G.; Wang X.; Wang P.; Ming H. Surface-Plasmon-Coupled Emission Microscopy with a Polarization Converter. Opt. Lett. 2013, 38, 736–738. 10.1364/OL.38.000736. [DOI] [PubMed] [Google Scholar]
- Zhang P.; Ma G.; Dong W.; Wan Z.; Wang S.; Tao N. Plasmonic Scattering Imaging of Single Proteins and Binding Kinetics. Nat. Methods 2020, 17, 1010–1017. 10.1038/s41592-020-0947-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang P.; Ma G.; Wan Z.; Wang S. Quantification of Single-Molecule Protein Binding Kinetics in Complex Media with Prism-Coupled Plasmonic Scattering Imaging. ACS Sens. 2021, 6, 1357–1366. 10.1021/acssensors.0c02729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu H.; Shan X.; Wang S.; Tao N. Achieving High Spatial Resolution Surface Plasmon Resonance Microscopy with Image Reconstruction. Anal. Chem. 2017, 89, 2704–2707. 10.1021/acs.analchem.6b05049. [DOI] [PubMed] [Google Scholar]
- Yang Y.; Shen G.; Wang H.; Li H.; Zhang T.; Tao N.; Ding X.; Yu H. Interferometric Plasmonic Imaging and Detection of Single Exosomes. Proc. Natl. Acad. Sci. U. S. A. 2018, 115, 10275–10280. 10.1073/pnas.1804548115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sung J.; Schnedermann C.; Ni L.; Sadhanala A.; Chen R. Y. S.; Cho C.; Priest L.; Lim J. M.; Kim H. K.; Monserrat B.; et al. Long-Range Ballistic Propagation of Carriers in Methylammonium Lead Iodide Perovskite Thin Films. Nat. Phys. 2020, 16, 171–176. 10.1038/s41567-019-0730-2. [DOI] [Google Scholar]
- Delor M.; Weaver H. L.; Yu Q. Q.; Ginsberg N. S. Imaging Material Functionality through Three-Dimensional Nanoscale Tracking of Energy Flow. Nat. Mater. 2020, 19, 56–62. 10.1038/s41563-019-0498-x. [DOI] [PubMed] [Google Scholar]
- Merryweather A. J.; Schnedermann C.; Jacquet Q.; Grey C. P.; Rao A. Operando Optical Tracking of Single-Particle Ion Dynamics and Phase Transitions in Batteries. Nature 2021, 594, 522–528. 10.1038/s41586-021-03584-2. [DOI] [PubMed] [Google Scholar]
- Bai Y.; Yin J.; Cheng J.-X. Bond-Selective Imaging by Optically Sensing the Mid-Infrared Photothermal Effect. Sci. Adv. 2021, 7, eabg1559 10.1126/sciadv.abg1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D.; Li C.; Zhang C.; Slipchenko M. N.; Eakins G.; Cheng J.-X. Depth-Resolved Mid-Infrared Photothermal Imaging of Living Cells and Organisms with Submicrometer Spatial Resolution. Sci. Adv. 2016, 2, e1600521 10.1126/sciadv.1600521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z.; Aleshire K.; Kuno M.; Hartland G. V. Super-Resolution Far-Field Infrared Imaging by Photothermal Heterodyne Imaging. J. Phys. Chem. B 2017, 121, 8838–8846. 10.1021/acs.jpcb.7b06065. [DOI] [PubMed] [Google Scholar]
- Bai Y.; Zhang D.; Lan L.; Huang Y.; Maize K.; Shakouri A.; Cheng J.-X. Ultrafast Chemical Imaging by Widefield Photothermal Sensing of Infrared Absorption. Sci. Adv. 2019, 5, eaav7127 10.1126/sciadv.aav7127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavlovetc I. M.; Podshivaylov E. A.; Chatterjee R.; Hartland G. V.; Frantsuzov P. A.; Kuno M. Infrared Photothermal Heterodyne Imaging: Contrast Mechanism and Detection Limits. J. Appl. Phys. 2020, 127, 165101. 10.1063/1.5142277. [DOI] [Google Scholar]
- Zhang Y.; Yurdakul C.; Devaux A. J.; Wang L.; Xu X. G.; Connor J. H.; Ünlü M. S.; Cheng J.-X. Vibrational Spectroscopic Detection of a Single Virus by Mid-Infrared Photothermal Microscopy. Anal. Chem. 2021, 93, 4100–4107. 10.1021/acs.analchem.0c05333. [DOI] [PMC free article] [PubMed] [Google Scholar]


















