You are on page 1of 13

Chemical Physics Impact 2 (2021) 100013

Contents lists available at ScienceDirect

Chemical Physics Impact


journal homepage: www.elsevier.com/locate/chphi

Viral particle imaging by super-resolution fluorescence microscopy


Stefania Castelletto a,∗, Alberto Boretti b
a
RMIT University, Melbourne, Victoria, Australia
b
Prince Mohammad Bin Fahad University, Al Khobar, Saudi Arabia

a r t i c l e i n f o a b s t r a c t

Keywords: Fluorescence microscopy provides a facile imaging methodology and potentials for imaging viruses in vivo. How-
Fluorescence microscopy ever, it is limited by the diffraction limit of light microscopy which is above the size of the virus particles. Flu-
Virus entry orescence super-resolution (FSR) microscopy circumvents the diffraction limit. These techniques, an extension
Super-resolution imaging
of fluorescence microscopy and alternative to electron microscopy, established over the past twenty years, have
permitted the visualization of some virus components. Here, we summarize the principles of different FSR tech-
niques and single virus tracking, and their current applications to virology studies, focusing on some limitations
impeding their widespread use in virus replication and infection mechanism studies in living cells. In the spe-
cific context of virology studies and live-cell imaging, the advantages and disadvantages of the most known FSR
methods are outlined with a focus on their current limitations such as labeling, imaging speed, photo-bleaching,
depth of imaging, and the current attempts to overcome them. Some of the recent demonstrations encompassing
the combination of different super-resolution (SR) approaches have provided answers to open questions as an
example in the human immunodeficiency virus replication and from these experiences, further progress can be
envisaged to reveal other viruses.

Introduction cell, to achieve the infection. They can be summarized in the following
steps whose details depend on the specific virus [1]:
Viruses also referred to as viral particles or virions are nanoscale
genetic entities that can replicate only within cells of plants, fungi, ani- 1. diffusion on the surface of the cell and binding to cells receptors or
mals, or humans. Viruses cores are made of a single nucleic acid strain host cells membrane;
(RNA or DNA) also referred to as viral genome within a capsid, enclosed 2. enter in the cell through endocytosis;
by a coat covered by proteins, sometimes enzymes, and in case of en- 3. viral membrane fusion (most enveloped viruses) or cellular mem-
veloped viruses, by a lipid membrane. The virus enclosure can vary and brane fusion (most non-enveloped viruses). Here the viral genome is
undergo assembly to initiate viral infection and replication. Outside a released in the cell;
living cell, viruses cannot reproduce but are however capable of infect- 4. replication of the viral genome and translation of additional viral
ing the cells and rely on the host cells for the completion of their life proteins required for new rounds of replication;
cycle. Retroviruses like HIV enter in hosts’ cells to reproduce by integrat- 5. assembly and packaging of the new viral genomes into virus parti-
ing their viral genome into the host genome, and their replication occurs cles;
along with the host genome replication during each cell cycle. This is not 6. budding where the new virus prepared to be released and ready to
the case with other viruses such as Hepatitis, Influenza, or Polio. In gen- infect new cells;
eral, the entry of a virus into a cell starts with attachment to receptors. 7. maturation.
This is then followed by conformational changes of viral proteins, and
The details of the human immunodeficiency virus (HIV) life cycle is
in case of non-enveloped viruses penetration-through cell membranes,
provided in Fig. 1 (a) and also summarized in ref. [2], while Fig. 1 (b)
or cases of enveloped viruses fusion with cellular membranes. Transfer
shows the six steps entry of the Influenza A virus (IVA) as studied in ref.
of viral genomes inside host cells eventually occurs.
[3]. Both viruses were further studied using super-resolution methods.
Virus components are generally much smaller and their identifica-
Cell and molecular biologists, virologists, and immunologists need
tion and visualization are critical in following the virus lifecycle. Virus
novel technologies to better understand the above mechanisms at the
lifecycle involves complex interactions between the virus and the host
single virus level that are specific to each virus. Visualization of the
way the virus enters the cell and transports the viral genome needed


Corresponding author.
E-mail addresses: stefania.castelletto@rmit.edu.au (S. Castelletto), a.a.boretti@gmail.com (A. Boretti).

https://doi.org/10.1016/j.chphi.2021.100013
Received 17 September 2020; Received in revised form 1 February 2021; Accepted 18 February 2021
2667-0224/© 2021 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Fig. 1. (a) Illustration shows the HIV replica-


tion cycle, beginning when HIV fuses with the
surface of the host cell, replication, assembly
and packaging, budding and maturation Credit:
NIAID (b) Sequential events during cells entry
for influenza A virus (IAV) obtained by con-
focal fluorescence and machine learning meth-
ods from [3]. Creative Commons Attribution Li-
cense.

for its reproduction is of paramount importance to viral studies, gener- tivirals. The ultimate quest is to image a single virus with high resolu-
ally studied via ensemble biochemistry providing a statistical approach. tion and to track single virus dynamics in living cells. For this purpose,
However, while a single virus can infect a cell, only a small percentage single-molecule fluorescence microscopy can reveal virus infections and
of them, for example for retroviruses as HIV that bind to the cell sur- biology that are inaccessible otherwise. A recent review has focused on
face, are eventually able to infect the host cell with replication of their the key mechanism in virus-cell interaction, virus cell entry, and trans-
genome [4]. There is therefore significant interest in imaging and track- port observed under various single-molecule fluorescence microscopy
ing of viruses at the single virus level to understand their life cycle. Most and associated SR techniques [1].
current studies on virus imaging are focused on uncovering the mecha- To this end, several methods have been deployed which can address
nism of assembly and cell interaction and infection and the single virus either resolution or tracking, either in ensemble measurements or at the
imaging can provide a novel understanding of these mechanisms. Single- single level, mostly in vitro. These methods are based mainly on electron
virus and single-molecule analyses can reveal which different steps and microscopy which provides enough resolution with however a lack of
host factors are required for a successful infection. The recent outbreak dynamic imaging and limited applicability to in vivo imaging due to the
of SARS-CoV-2 has brought even more attention to the need for imag- vacuum operation conditions or complex sample preparation. Later fluo-
ing virus with the desired resolution to uncover the specific mechanisms rescence microscopy, either wide-field or scanning confocal microscopy,
associated with the cell infection process [5]. has been applied to the virus, however, these methods lack enough reso-
Viruses such as HIV, influenza, and SARS-CoV-2 are approximately lution, while having the possibility of single virus imaging and tracking
100 nm in diameter, thus considerably smaller than cells even if some in principle in vivo. More recently fluorescence super-resolution (FSR)
of them can reach the size of bacterial cells [6]. Their constituents methods have been applied to virus imaging with the potential of in vivo
are therefore much smaller than 100 nm. Due to this small size super- applications. At present, none of the imaging and tracking techniques
resolution (SR) imaging techniques, as well as high localization for their can accomplish all the above requirements. However, by learning from
tracking, is necessary. A better knowledge of the mechanisms of virus the current demonstrations of FSR techniques in specific viruses, further
assembly and infection is needed for prophylaxis and treatment by an- development can be envisioned to accomplish some of these quests.

2
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

The virology world’s major quest of observing in vivo and real-time FSR principles and techniques
the interaction of the nanometric scale virions with the molecular cell
biology is currently barely within reach. This is because of the differ- FSR microscopy techniques recently established [14,18], com-
ent spatial scales involved, the time scale within which current high- prise among the most notable, single-molecule switching or localiza-
resolution imaging can be achieved, and the difficulty to image for a tion microscopy (SMSM) divided into Photo-Activation Localization
long time due to either methods’ toxicity responsible for cells’ death or Microscopy (PALM) [19–21] and Stochastic Optical Reconstruction
tags photo-bleaching. Microscopy (STORM) [22,23], and stimulated emission depletion mi-
Regardless of the possibility of live-cell imaging studies, most to-date croscopy (STED) [24]. As a common feature, these methods permit to
virus studies using FSR microscopy and single virus tracking have been achieve spatial resolution below 100 nm, enabling the imaging of viruses
carried out only on fixed samples. For virology research in vivo, these for the examination of their architectural details and their interactions
technologies necessitate in addition to a higher resolution than current with the cell constituents throughout virus replication, spread, and in-
capabilities for in vivo imaging, also low photo-bleaching and photo- fection. These methods have permitted to resolve images better than 20
toxicity, higher imaging speed, and the possibility to achieve imaging nm and achieve 10 nm localization accuracy. While high resolution can
of the sample with depth penetration and specific labeling. In particu- be achieved with these methods, they suffer from slow acquisition time
lar, the acquisition of cells and virus dynamics in the time scale of ms, and/or post-processing of images, as well in some cases of photo-toxicity
requires high imaging speed, while their interaction should allow long or photo-bleaching of fluorescent tags that make volumetric time elapse
tracking times. This is everything but simple to achieve due to short life imaging of living samples impractical.
fluorescent labels and photo-toxicity leading to cells death. Principles of the most prominent FSR microscopy methods with their
This paper focus on the latest developments FSR microscopy has typical achievable resolution are provided in Fig. 2.A, where the inten-
achieved and may perform in the future regarding viral particle stud- sity distribution of 550 nm laser beam through a commonly used oil im-
ies. mersion objective (Obj) with numerical aperture NA = 1.4, is displayed
Here, we briefly summarize the historical approaches to image in the focal point. The beam intensity distribution at the focal point
viruses and the latest attempts by using the most popular FSR meth- in the lateral and axial directions are ~250 nm and ~600 nm, respec-
ods. We then focus on their current achievements and limitations with tively, in a confocal or wide-field microscope with schematics shown in
regards to open problems associated with these methods impeding to Fig. 2.B. While scanning confocal fluorescence microscopy using point
reach in vivo single virus imaging [7]. In particular, the development detectors and a pinhole for read-out to improve spatial resolution and
of FSR methods for virus imaging, while have reached a high level of reduce the contribution from the sample background out of focus, wide-
resolution ideal for viruses, has been impacted by common open limi- field microscopy does not require scanning and has generally a larger
tations such as accurate labeling, imaging speed, sample depth penetra- field of view.
tion, photo-bleaching, and photo-toxicity. By using SIM and similar techniques based on illumination with
structured multifocal points, or Airyscan detection mode [25], it is pos-
Fluorescence microscopy methods for viral particle imaging sible to achieve a spatial resolution of 100 nm and 300 nm in the lat-
eral and axial directions, respectively, accounting for an improvement
Due to the nanometric virus size, early imaging methods and cur- of a factor of 2 versus conventional confocal or wide-field microscopes.
rently commonly used methods to achieve static images of viruses, are These approaches use the variation of light patterns in the detection
based on conventional electron microscopy such as scanning electron channel to reconstruct the image (Fig. 2.C). A series of images from the
microscopy (SEM) and transmission electron microscopy (TEM) [8], CCD camera is collected by turning the illumination structure and from
with viruses, imaged emerging from cultured cells in the laboratory. these images analysis, it is possible to recreate the final image with high-
However, these methods, while they can provide high resolution (sub resolution information [14]. In ref. [26] 3D-SIM permitted a two-fold
nanometric) in the imaging, are onerous from the sample preparation increase in resolution in all three dimensions of the Hendra virus (HeV)
point of view, can preclude live imaging, and have limited opportunity in fixed infected primary bovine, porcine aortic endothelial cells, and
for molecular identification [9]. Other methods such as atomic force mi- bat-derived cell lines. Here the HeV M and G proteins structures were
croscopy [10] or optical tweezers [11] can be employed to study prop- confirmed as being around HeV virion and SR imaging demonstrated
erties of the virus at the nanoscale, however with limited cases applica- for the first time sub-virion imaging of paramyxovirus proteins and the
bility and yet far from common. respective localisation of HeV G, M and N proteins within the viruses.
Fluorescence microscopy [12] is instead more commonly used and Recently using SIM and image reconstructing algorithm combined with
it has permitted virologists to study the virions’ interaction with cell SEM, Zika virus main stages of its assembly from viroplasm formation to
surface while entering the cell, using wide-field and scanning confocal the viral release in mammalian cells were studied [27]. 3D-SIM has been
fluorescence microscopy due to the relatively fast and easy methodol- used to study vaccinia virus replication resolving the virus morphologies
ogy. On the other hand, the resolution of a fluorescence microscope is at the scale of single virus particles [28].
𝜆
physically limited by the Abbe diffraction limit [13] given by 2 𝑁𝐴 ~ Total Internal Reflection Fluorescence microscopy (TIRF) [29], con-
190 in the plane of focus (xy) of an oil immersion objective with nu- sists of the formation of an induced evanescent wave at the interface
merical aperture NA=1.4, excited with a laser with wavelength of 532 of two media having different refractive indices, generally in a limited
nm and by 1.𝑁 4𝑛𝜆
𝐴2
~ 570 nm along the axial direction (in-depth) of a sample region close to the interface area between the sample and a
medium of refractive index n=1.5 [14]. The actual resolution in the glass coverslip or tissue culture container. In this case, the excitation
non-ideal experimental conditions is often even lower [15]. This type of beam is reflected totally at this interface, reducing the energy at a sam-
resolution is not enough to image a virus without obtaining a blurred ple region of interest with thus minimal background. This can reduce
image. the excitation power in axial z-direction thus reducing background and
Conventional wide-field and laser scanning confocal fluorescence permitting to achieve ~ 100 nm resolution in the axial direction [30].
microscopes principles have been developed by introducing some struc- TIRF combined with SIM has been implemented and can provide a good
tural optical modification to improve their spatial resolution. Structured spatio-temporal resolution, remaining a facile method, retaining versa-
illumination (SIM) [16,17] achieved improvement in axial resolution in tility, and permitting to achieve 3D live-cell imaging capability by using
the range of 100–150 nm. These approaches are quite simple and as conventional microscopy fluorophores. Therefore, it is considered the
such still offer the advantages of direct applicability, using convention- best method for studies requiring at the most 100 nm resolution, while
ally prepared fluorescent probes and samples. However, due to their still not enough in areas such as virus research, requiring less than 100 nm
limited optical resolution, their use is not wide-spread in virus research. resolution.

3
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Fig. 2. A. Lateral or in-plane (x-y) and ax-


ial (z-direction, depth) resolution of various
imaging schemes. Schematics of B. Confocal
and wide-field combined, C SIM, D STED, and
E 3D STORM [33]. EMCCD is an electron-
multiplied charge-coupled device used to im-
age low brightness fluorophores for higher
imaging resolution.

Fig. 3. Rotavirus RRV-infected MA104 cells


were fixed and processed for transmission
electron microscopy or immunofluorescence
microscopy. (A) Transmission electron mi-
croscopy of a VP (identified by the dotted white
ellipse). (B) Diffraction-limited image of VPs
(white arrows). (C) 3B-SR image reconstructed
from B. (D–E) 3B-SR images of individual VPs
labeled with different antibodies. Image from
ref. [32]. Creative Commons CC BY license. cre-
ativecommons.org/licenses/by/4.0/.

Interestingly by combining TIRF microscope with a Bayesian analysis their insight have been recently reviewed in ref. [14,38,39] and a re-
[31] of the stochastic blinking and bleaching (3B SR) of Alexa Fluor cent perspective on the use of FSR microscopy for HIV can be found
488 dye, a recent study [32] shows an achieved resolution of 50 nm in ref. [2]. These studies together with examples applied to Rotavirus
in imaging the viroplasms (VPs), cytoplasmic electron-dense inclusion [32,40], Hendra virus [26], Vaccinia Virus [28], VSV-like particles [41],
where the Rotavirus genome replication and assembly occur (Fig. 3). Influenza A virus (IAV) [42-44], ZIKA virus [27], Herpes simplex virus
This showed the structural organization of seven viral proteins within [45], Ebola virus [46], while in still limited demonstrations, illustrate
and around the VPs organized in five concentric layers. the journey of SR imaging to bring a better understanding of cell-virus
Sub-diffraction resolution below 100 nm attained in fluorescence mi- biological processes and their dynamics with nanoscale resolution.
croscopy relies on fluorescent labels being switched “on” and “off” be- Details on the techniques applied for the specific viruses and major
tween so-called “bright” and “dark” states either deterministically or insights on the virus interaction with the cells will be discussed in the
stochastically. This switching mechanism permits to fluoresce to only following sections, while we summarise the main or more recent ad-
a small subset of all labels in the focal spot, thus, to be individually ditional virus studies, methods, and finding using FSR or single virus
distinguished and localized. Their imaging and localization contribute tracking in Table 1.
to a reconstructed image defeating the light diffraction limit [34]. The We discuss some common fluorescent tags used in viral imaging and
underlining difference between techniques relies on the type of switch- other solid-state tags. We then focus on the application of these tech-
ing mechanism or localization, of either deterministic or stochastic ori- niques for viral particle imaging, while reminding the reader of more
gin. In a deterministic approach the fluorophore from its excited state specific details of the techniques to other reviews [14,61]. For a more
is forced into the dark state by using a dual excitation beam as in STED extensive summary of the many other techniques principle of operation
microscopy [24] and in the Reversible Saturable Optical (Fluorescence) and schematics see for example ref. [62].
Transition (RESOLFT) microscopy [35,36]. In the first method, the dark
state is a ground state while in the second is a long-lived metastable Fluorescent tags in viral particle imaging
state or intersystem crossing state.
To date, only a few demonstrations of the application of the above One of the key issues of fluorescence microscopy-based methods ap-
FSR methods to study the bio-functionalities of viruses even in fixed plied to the virus is the labeling or fluorescence tags. In general, with
cells can be accounted for, with the principal focus been the human im- all the above FSR approaches significant attention must be taken in the
munodeficiency (HIV) virus [37]. FSR microscopy studies of HIV and labeling of the virus by fluorescent markers or tags if compared with

4
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Table 1
Summary of specific referenced virus studies, FSR or single virus tracking methods used, and key findings.

Virus type Methods Key Findings Refs.

HIV-1 STED Dynamics imaging of Gag protein and Env during virus [47,48]
maturation.
HIV-1 STED-FCS Study of the mobility of the proteins on the surface of [49]
individual HIV-1 particles in live-cells context.
HIV-1 dSTORM-TIRF 2D molecular distribution of the major structural proteins of [50-53]
the HIV-1 after infection. Colocalization (Env and Gag) at the
HeLa cell membrane for the virus assembly
SERINC5 and anti-retroviral factor mechanism study.
HIV-1 PALM and TIRF HIV assembly and budding studies at the plasma membrane [54]
of CD4+ T cells
HSV-1 dSTORM Localized the protein layers on the virus in infected cells [55]
HSV-1 STED Identify proteins for processes of cell attachment and [56]
membrane fusion.
Nipah Virus dSTORM Reveal virus assembly model [57]
Rotavirus TIRF combined with Study of replication and assembly [32]
Image reconstructing
algorithms
Influenza Virus A Single virus tracking IAV virions encapsulating quantum dots are tracked in [43,58]
real-time, dynamic of the uncoating is revealed in real-time.
Influenza Virus A dSTORM Virus matrix protein localization [44]
Influenza Virus A STED Virus events trafficking in human dendritic cells after [59]
internalization
Ebola Virus Single Virus Tracking Virus internalization [46]
Zika Virus 3D-SIM plus image Main stages of virus assembly and release in mammalian [27]
reconstructing cells
algorithm and SEM
Human respiratory STED High-resolution images of the virus [60]
syncytial virus

confocal microscopy. The size of the fluorescent tags is of paramount im- Fluorescence proteins have been advanced for virus labeling appli-
portance to directly target the biological structures of interest and track cations using conventional microscopy and they can be implemented for
cellular processes in specific locations without introducing artifacts and live-cell FSR imaging. Other organic-dye tagging such as tetracysteine
interfere with the virus and cells’ functionality. Besides, they must have (TC) tag, CLIP-tag, SNAP-tag, or artificial amino acids and click chem-
photo-physics properties that are compatible with the microscopy and istry are viable labeling methods to accomplish FSR imaging studies of
FSR principles, and ideally, they should not photo-bleach to provide ac- virus replication cycle in living-cell [66].
cess to an extended time for tracking. For FSR tagging each virus compo- Standard commercially available Alexa Fluor and ATTO dyes [67],
nent with high molecular specificity and efficiency is essential to achieve which cover the visible wavelength range, are switched reversibly be-
relevant information in their dynamics during the virus lifecycle. Fur- tween an “on” and an “off” state with the addition of thiol‐containing
thermore, depending on the process associated to each viral component reducing agents which are used to efficiently induce a relatively stable
to be imaged, fluorophores with distinct fluorescence properties, such as non-fluorescent state [67]. These small organic fluorophores are rele-
brightness, fluorescence lifetime, photostability, emission wavelength, vant for super‐resolution imaging in living cells.
photo-switching capabilities have to be specifically tailored. For exam- Solid-state nanoparticles such as QDs has been more recently applied
ple, some virus components such as proteins and the lipid envelope can for SVT [7,43,68,69] also in combination with other labeling methods
be labeled using synthetic fluorophores as described also in ref. [63]. [70], suggesting that solid-state labeling is the latest trend in the single
Specifically, several tags have been used for fluorescence microscopy virus tracking [43,58].
of viruses such as covalent labeling, intercalating and lipophilic dyes, Other solid-state nanoprobes have been proved efficient for FSR
for single virus imaging, fluorescent proteins, fluorescent nanoparticles imaging. For example, nanodiamonds (NDs) also can bind to viruses,
such as quantum dots (QDs), and metal nanoparticles, as discussed in such as hepatitis B or C, from blood plasma isolated from infected pa-
detail in ref. [7]. So far, the virions have been conjugated mostly with tients [71]. As NDs contain a nitrogen-vacancy (NV) color centers that
organic fluorophores or genetically combined to a green fluorescent pro- can be used as a fluorescent tag in the near-infrared without photo-
tein (GFP). The fluorescence can be induced on the virion by the so- bleaching, they can be bonded to viruses and used are FSR solid-
called split-GFP technology consisting of an engineered particle with a state probes. FSR imaging using NDs has been already demonstrated
GFP segment bond to a protein of interest associated with another GFP in several examples with 20 nm resolution using SMSM [72,73], spin-
segment [64]. A recent comprehensive review of the type of fluorescent- dependent fluorescence [74], STED [75], SPIN-RESOLFT [76] as well
labeled viruses and fluorescence microscopy methods to image them so as the paramagnetic properties of NV centers can be used to perform
far is reported in ref. [6]. spin nanoscopy [73], magnetic imaging combined with FSR [77], and
Antibodies are considered large fluorescent tags whose size can bias spin-STORM [78]. NDs containing NV is also successfully applied to
the super-resolved image and therefore introduce errors in the localiza- two-photon microscopy with adaptive optics [79]. STED microscopy
tion of the tagged molecules. Smaller tags such as nano-bodies or click has achieved the best resolution using NV in diamond [80], and SMSM
chemistry are therefore essential for FSR microscopy applications [65]. techniques benefit from NDs as labels due to lower photo-toxicity and
Live-cell FSR microscopy studies of viruses appear to be even more sus- better photo-stability not requiring near UV excitation to induce photo-
ceptible to issues with labeling with stricter requirements of minimal switching mechanism. The current limitation to use NDs in FSR imag-
interaction with the virus structure and functionalities. Antibodies or ing of biological systems such as virus is the relatively large and variable
nano-bodies for fluorescence tagging have limited possibility in live-cell size of NDs (~35-50nm), while other applications as fluorescent labeling
imaging because they can only be applied to study external surfaces of of the biological sample have been established [81]. Other fluorescent
the viruses and cells. probes based on solid states nanomaterials have been recently applied

5
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

to SMSM such as silicon carbide nanoparticles [82] and boron nitride


flakes [83], the first having the possibility to achieve dual-color photo-
switching using 640 nm beam, thus reducing further photo-toxicity and
photo-bleaching. Both materials can be fabricated in < 8nm size, while
still maintaining their fluorescence brightness. However, none of the
latest proposed novel fluorescent solid-state labels for single-molecule
imaging, have been applied to viral imaging as they are at a very early
stage of development.
The tagging of the viral genome of the virus appears much more
difficult and so far studied mostly for HIV by fusing a fluorescent
protein to an RNA-binding protein or by using fluorescence in situ
hybridization [2].

STED microscopy and fluorescence correlation spectroscopy in


viral imaging

STED microscopy [84] (Fig. 2D) relies on using generally a green


laser excitation to induce spontaneous decay of the fluorophores from
their excited state to the ground state by emitting fluorescence photons.
A red-shifted laser excitation (known as STED laser) is applied to the
excited fluorophore to induce stimulated emission to the ground state
occurring without photon emission in the spectral region of interest. The
STED beam is engineered via a phase modulator to achieve in the focal
point a “donut-shaped” intensity distribution, corresponding to a cen-
tral intensity minimum, where the spontaneous fluorescence is depleted
everywhere except at the center. This permits localization of the fluo-
rophore with a point spread function much smaller than the focal spot of
the excitation beam, which is otherwise diffraction-limited. The resolu-
tion is given by the efficiency of the fluorescence depletion depending on
the intensity of the STED laser. The resolution can be tuned by adjusting
the STED laser shape and intensity, achieving a resolution < 50–60 nm in
the plane direction in cellular imaging [85,86] and < 100 nm resolution
in the axial direction in biological samples [87]. The advantage of STED
microscopy is the direct creation of the image in real-time with a simple
acquisition process, without the need for image frame post-processing,
which may induce potential post-processing image artifacts. However,
achieving high resolution and high speed comes at the expense of photo-
toxicity. When exposing samples with high laser intensities in the ~ GW
cm−2 as with the STED-beam, radicals or singlet oxygen can be gener-
ated causing photo-bleaching and photo-toxicity in living systems with
subsequent cell death. Living cell imaging with STED has been how-
ever achieved [88-90] by optimising the sample preparation protocols
and using fast beam-scanning methods. Nevertheless, STED microscopy
even if it can be used for fast live and fixed cells studies, due to the in-
evitable still too high laser power required, it is not suitable for a long
Fig. 4. a Live confocal imaging of HIV-1 virus in a 2 × 2 μm2 imaging frame
period of live-cell imaging. using the GFP labeling the viral protein Vpr, GFP-Vpr (green). The scale bar
High resolution can be combined also with high imaging speed in is 200 nm. In red the image scanning line locating the position of the virus.
STED microscopy. As STED is based on point by point laser beam scan- b, c The fluorescence intensity for GFP-Vpr (green) in confocal operation and
ning, the imaged field of view impacts its imaging speed with smaller Env (orange) in STED operation on mature HIV-1 particle. The fluorescence
image implying a faster acquisition time. The maximum temporal reso- intensity is scanned along with the red line position (x-axis) of the virus and
lution of STED-microscopy of 5–10 ms was achieved in the investigation versus time (y-axis), respectively. Scale bars are x-axis (white) = 200 nm, y-axis
of HIV-1 (the most virulent of the two HIV strains) acceptance into HeLa (grey) = 4.4 ms. d Mapping of the virus protein Env position and time as acquired
cells. Such high temporal resolution was achieved by reducing the ac- in c, in terms of the correlation function, showing the possibility to track the
intensity correlation of the proteins at the virus position with super-resolution.
quisition time by employing ultrafast laser-scanners on small areas of
e Normalised autocorrelation curves G(𝜏) of Env diffusion (grey and black lines)
interest, namely a reduced field of view [91]. Here STED was modified
for a specific position on the scanned line for the mature (red), immature (blue),
to be able to image the cellular uptake of viral particles with 5- to 10-ms and fixed HIV-1 particles (purple). Figure from ref. [95]. Creative Commons CC
temporal resolution. BY license. creativecommons.org/licenses/by/4.0/.
In general, for all FSR methods included STED, to achieve sub-
diffraction resolution on a given field of view a long acquisition time is
needed. The field of view is however important in imaging virus interac- lease of individual proteins. The structural polyproteins Gag of HIV-1
tions with cells and it cannot be sacrificed. Currently, STED provides the are essential players for the assembly, release, and maturation of virus
best imaging speed of the fluorescence-based SR techniques however at particles and are considered for anti-viral therapy [92]. The incomplete
the expense of a limited field of view. Several studies using STED were Gag lattice of immature virions was distinguishable from the condensed
performed in HIV-1. distribution of mature protein subunits using STED, as compared to elec-
For HIV-1 to achieve a morphologically mature fully infectious virus tron tomography [93]. The maturation is a critical step to understand
particle status, it undergoes a process called maturation with the re- the HIV-1 replication cycle and to develop anti-HIV-1 therapy. The HIV-

6
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

1 maturation and how the virus evolves into a lipid environment were the input IAV nucleoprotein, early and late endosomal compartments,
studied using STED. The study permitted visualization of the protein and dendritic cells membrane/cytosol.
on the surface of individual HIV-1 particles. The first visualization of Human respiratory syncytial virus (RSV) forms pleomorphic virus
the viral envelope (Env, the trimeric viral envelope surface glycopro- particles made of long filaments about 100 nm in diameter and up to
tein) glycoprotein distribution on the surface- with features of 40nm- about 10 𝜇m in length. RSV affects human lung epithelial cells induc-
of individual HIV-1 particles was achieved with STED using anti-gp120 ing filopodia, cellular protrusions that extend to neighboring uninfected
antibodies as reporters [47,48]. This study revealed that the maturation cells. SR microscopy techniques are needed to visualize the interactions
was induced by clustering on the virus surface of the Env proteins that between RSV infected cells and virus particles. In ref. [60], STED mi-
depended on the Gag-interacting Env tail [47]. STED was able to visual- croscopy combined with software providing image deconvolution to cor-
ize these proteins on the virus’s surface in different stages as it was not rect for optical distortion has revealed filamentous RSV particles along
possible with confocal fluorescent microscopy [47]. Previous live-cell these filopodia, providing clues that filopodia facilitates RSV spreading
and STED studies on HIV-1 were limited by requiring antibody labeling from cell to cell. Specific dyes were used to prepare fixated cells with
for Env detection. In [94] a bio-orthogonal amino acid is combined with RVS and STED permitted to discern and enumerate the RVS particles
click chemistry for site-specific and minimally invasive labeling. attached to filopodia.
STED was combined with another method called Fluorescence Corre- For the Herpes Simplex Virus 1 (HSV-1) the processes of cell attach-
lation Spectroscopy (FCS) [95], based on the correlation analysis of the ment and membrane fusion involve many different envelope glycopro-
fluorescence intensity fluctuations of the tagged molecules over time teins (called gB, gC, gD, gH, and gL) which bind to cellular receptors.
while they diffuse. From the correlation of their intensity fluctuations, Using STED it has been shown that most of these proteins are distributed
it is possible to determine the molecular transit times and their diffu- evenly round purified virions, while gD localizes essentially as clusters
sion coefficient. FCS measurements can also enable the determination that are distinct from gB and gH/gL while it localizes with other gly-
of physical parameters governing cellular mechanisms and dynamics, es- coproteins on cell-bound particles. This redistribution of gD upon cell
pecially when high sensitivity and fast dynamic resolution are required attachment could be responsible for the activation of membrane fusion
[96]. FCS when combined with STED (known as STED–FCS) can achieve [56].
the determination of the molecular diffusion with high spatial localiza-
tion at the level of an individual molecule. STED-FCS also improve tem- Single-molecule switching microscopy
poral resolution [97] to make it more applicable to virus study.
STED-FCS permitted to study the lipid envelope surrounding some Other FSR techniques use a stochastic intensity switching, resulting
viruses in detail for example, in a live-cell context. Specifically, STED- in fluorescence intermittency of fluorescent molecules or other probes,
FCS has been utilized on HIV-1 and permitted to study how HIV-1 in the entire field of view of a wide-field microscope. Using imag-
evolves into a lipid environment, allowing the virus to assemble in the ing reconstruction algorithms, the precise localization of the probes is
plasma membranes [95]. The study permitted to determine the mobility achieved. Single-Molecule Switching Microscopy (SMSM) is the general
of the protein on the surface of individual HIV-1 particles [95]. Specif- term used to group all the techniques based on this approach, which
ically, it was confirmed that during its assembly, HIV-1 incorporates includes PALM [21,41] and STORM [22,33] as well as some variations
copies of Env1, 2 (the trimeric viral envelope surface glycoprotein), and of them such as Super-resolution Optical Fluctuation Imaging (SOFI)
the main structural protein Gag3 (a polyprotein initially accumulating [102].
into immature virus particles) [98]. To achieve a morphologically ma- SMSM-methods are usually based on wide-field microscopy with
ture fully infectious virus particle, maturation with the release of indi- read-out achieved by EMCC detection (Fig. 2 B). Here a sequence of
vidual proteins occurs. 100 up to 10,000 individual camera frames are taken, imaging only a
In Fig. 4 STED-FCS is applied to measure the autocorrelation curves limited number and different subsets of individual isolated fluorescent
G(𝜏) of Env diffusion on HIV-1 particles for different virus phases, named labels each time, as they are stochastically switched-on for each subse-
mature, immature, and mature fixed HIV-1 virus conditions. It was quent camera frame. The sub-100 nm resolution image is reconstructed
found that Env undergoes an increase in mobility due to maturation. from these many camera frames by determining the spatial positions of
This technique has provided appreciable information on the protein’s the individual fluorescent molecules from their intensity “on” and “off”
assembly dynamics and fusion at virus surfaces in vivo. states temporal dynamics. The various methods differ owing to the use
The membrane-proximal external region (MPER) of the HIV-1 Env of different fluorophores photo-physics to achieve their stochastic on-off
has been identified as a potential vaccine target [99]. The MPER acces- switching. PALM employs the fluorescence activation, using specific ex-
sibility is limited making its native structure poorly understood, as such citation lasers, of so-called “photo-activable” fluorescent labels, brought
different and exclusive models are explaining the molecular mechanism to an “on” state by the laser excitation; their emission then undergoes a
of MPER used by broadly neutralizing antibodies (bnAbs) to recognize subsequent photo-bleaching. Specifically, PALM uses photo-activatable
it. Ref. [100] studies the accessibility of antibodies to the native Env GFP (PA‐GFP) or mEOS as labels, which are genetically engineered fluo-
MPER on single HIV virions using STED. rescent proteins. PA‐GFP is initially non-fluorescent, while it is activated
Broadly neutralizing antibodies 10E8 and 4E10 Fabs were conju- in a fluorescent state using a pulse of 405‐nm laser light. By regulating
gated with the fluorescent probe Abberior STAR RED (also known as the laser intensity, only subsets of PA‐GFP molecules become activated
KK114) to analyze MPER accessibility through STED microscopy in the and are imaged and resolved using 561 nm laser, while the other re-
native Env complex. mains dark. STORM instead originally utilized stochastic fluorescence
STED imaging of fluorescently labeled Fabs reveals a common pat- transitions of organic dye [22]. In STORM the switchable fluorophore
tern of native Env recognition for HIV-1 antibodies targeting MPER or is a Cy3–Cy5 dye pair attached to DNA or protein. As opposite to PALM
the surface subunit gp120. Fig. 5 (a) presents the model of HIV viri- where photo-bleaching occurs, in the STORM method, the fluorescence
ons, and Fig. 5 (b,c) the visualization of these HIV virions by STED mi- is brighter and each fluorescent molecule can be cycled hundreds of
croscopy super-resolution. times before the irreversible bleaching occurs [22]. Later, studies found
The transport, acidification, and fusion of single influenza virus A that conventional dye fluorophores could be photo-switched without
(IVA) in living cells were studied by using fluorescence microscopy [3], the need for an activator fluorophore [23,103] enabling the application
which permitted to dissect individual stages of the viral entry pathway of existing antibody and nucleic acid labels in SMSM. Direct STORM
[101]. More recently three-color STED microscopy was applied to IVA to (d-STORM) relies on the intrinsic cycling of fluorophores between “on”
visualize early IAV events trafficking in human dendritic cells following and “off” states without the need for a secondary activator dye as in the
the internalization of virus particles [59]. In particular, it was visualized original STORM. In a more recently developed direct STORM (dSTORM)

7
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Fig. 5. (a) Model for MPER accessibility within native Env complexes based on Cryo-EM reconstructions. In the Top Contours showing the exposure of the MPER
helix in native Env (green) to 10E8 compared to no 10E8. Bottom cartoons showing the different Env components, and positions of the neutralizing epitopes for
bnAbs used PGT145 (V1/V2), VRC01, and b12 (CD4bs) and 10E8/4E10 (MPER). Image from [100]. This open-access article is distributed under Creative Commons
Attribution License 4.0(CC BY). (b) MPER Env site visualized by the signal of 10E8-KK114 bound to HIV-1 virions acquired in confocal (top) or STED modes (bottom).
(c) STED images of 2G12, 10E8-KK114, 4E10-KK114, and Annexin V-ATTO 647 bound to HIV-1 virions (top) and corresponding pixel areas (bottom). Image from
[100]. This open-access article is distributed under Creative Commons Attribution License 4.0(CC BY).

[104], conventional organic fluorophores, such as Cy5 and Alexa 647 In ref. [55], dSTORM was used to determine the position of pro-
[23] are used for reversible photo-switching without requiring an acti- tein layers within individual Herpes simplex virus type-1 (HSV-1) par-
vator molecule, this further improves the quality of signals. ticles. The envelope-anchored glycoproteins were discerned from tegu-
SMSM provides a higher resolution of 10-20 nm localization preci- ment proteins, both in purified virions and in virions present in infected
sions compared to STED. SMSM is also commonly paired with TIRF. cells.
SMSM-based imaging has been further improved by optimization of the Several studies using dSTORM and PALM as well as Single Particle
fluorophores photo-switching methods [23,105,106] by using as exam- Tracking PALM were performed on the HIV-1 virus revealing its proteins
ple multicolor imaging [107,108,109] and by introducing various 3D component, assembly, maturation, and replication cycles, whose details
SMSM approaches [33,110]. are summarized in ref. [38,98].
SMSM makes use of laser intensities in the ~kW cm−2 , much lower In ref. [50], dSTORM is combined with standard immunocytochem-
compared to STED, however, for photo-switching, it is often required istry and TIRF microscopy to quantify the 2D molecular distribution of
UV laser irradiation which can cause pronounced photo-toxicity. the major structural proteins of the HIV-1. HIV-1 is imaged before and
Currently, SMSM approaches encompass higher image resolution after the infection of lymphoid cells, after fixing the infected cells. It was
and lower photo-toxicity if it is compared to the other methods, how- found that after HIV-1 infection, the HIV-1 capsid proteins undergo re-
ever, the downfall is the requirement for acquisition of many camera arrangement. HIV-1 virus contained the GFP-viral protein (GFP-Vpr), to
frames and the use of post-processing algorithms to create a final highly allow visualizing particles with conventional fluorescence microscopy
resolved image which limits the temporal speed. SMSM techniques in [50]. Subsequently, dSTORM and TIRF using multicolor tags also re-
general feature a very high spatial resolution at the expense of reduced vealed information on protein colocalization (Env and Gag) at the HeLa
imaging speed due to the acquisition of thousands of photo-switching cell membrane for the virus assembly [52] as well as recruitment of
cycles necessary to build up the final image. tetherin clusters to Gag assembly sites at the membrane of HeLa cells
The long acquisition can introduce bias such as drift, which requires [51].
fiducial markers and correction to avoid image artifact. Long acquisition Recently, 2D and 3D dSTORM [53] revealed the mechanism by
times more importantly introduce an upper limit to the time resolution which Serine incorporator protein 5 (SERINC5), an anti-retroviral fac-
and speed when it is needed to resolve live-cell dynamics or to achieve tor, inhibits the Env mediated HIV-1 virus fusion with target cells by
real-time imaging of viruses infecting the cell as an example. Optimized imaging the distribution of Env on a single virus. It was first confirmed
image acquisition and processing protocols [111,112] have been devel- that the Env glycoproteins form clusters on the surface of mature virions
oped to mitigate these issues. Due to a several seconds typical acquisi- [47] and the SERINC5 disrupted Env clusters without affecting the over-
tion time associated with SMLM, the technique appears less suitable for all Env content, suggesting an indirect effect of SERINC5 on Env cluster
live-cell imaging while it is better applicable to fixed samples requiring formation. Recently dSTORM (based on commercial Nikon N-STORM)
the highest possible, molecular level resolution. has been used to study a matrix protein localization at the base of IAV
SMLM time resolution has been improved to reach 0.5 s in using sin- filaments, emerging from the membrane of infected stained cells [44].
gle excitation in a 2D imaging in cell environment [111], even if at a Correlative fluorescence light microscopy permits a combination of
reduced spatial resolution of 25 nm; while a 3D spatial resolution of ~30 SEM and FSR and it has been applied to the influenza virus [42], where
nm in the lateral directions and ~50 nm in the axial direction could be 3D STORM and SEM are combined (Fig. 6).
achieved with a time resolution of 1-2 s. This temporal speed is not still PALM microscopy has also demonstrated the ability to observe the
enough to capture live imaging of virus-cell molecular interactions as- virus assembly at the host cells with nanoscale resolution. In particular,
sociated with molecular diffusion and clustering dynamics, which occur it has been applied to immune system living host CD4+ T cells where
within a time scale of milliseconds at nanometer distances. dSTORM us- HIV assembly and budding at the plasma membrane of the cells was
ing small organic fluorophores with high photostability permits longer studied [54,98]. It has been implemented so-called live PALM, consist-
acquisition time thus can achieve a lateral resolution of 15–20 nm [67]. ing of combining PALM and TIRF-microscopy, with analysis methods

8
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Fig. 6. Correlated 3D STORM and SEM images


of individual filamentous influenza viruses (A)
Process to achieve correlative images. (B) Cor-
related images of STORM, overlaid image, and
SEM image of the virus. (C) Magnified images
of the selected areas in (B). (D) Transverse lo-
calisations of the selected area in (B). (E) Nor-
malized photon numbers per switching event.
(F) Cross-correlation between STORM and SEM
images. (G) Correlative two-color STORM and
SEM images of virus filaments with two differ-
ent immuno-labeling STORM image, Overlaid
image, SEM image. Scale bars are 500 nm De-
tails are reported in ref. [42]. This is an open-
access article distributed under the terms of the
Creative Commons Attribution License, which
permits unrestricted use, distribution, and re-
production in any medium, provided the origi-
nal author and source are credited.

Fig. 7. PALM and TIRF Imaging of mem-


brane Gag cluster sizes and densities at the
T-cell surface of fixed Jurkat T cells that ex-
press wild type (WT) Gag, NL4.3ΔPolΔEnv
Gag, WM, MACASP1, or Δp6. Purified virus-
like particles are on the left upper panel from
WT Gag productive cells used as a size ref-
erence. The major image with a 2 𝜇m scale
has a zoom of a selected area with a 1 𝜇m
scale bar. Image adapted from ref. [54]. Cre-
ative Commons CC BY license. creativecom-
mons.org/licenses/by/4.0/.

based on hidden Markov statistical analyses and Bayesian inference. protein mEOS2 to form Gag(i)mEOS2, PALM imaging was achieved. In
This approach has permitted to achieve a description of the virus for- Fig. 7 images using Live PALM show HIV-1 with various Gag proteins
mation at the plasma membrane of individual cells, with 50 nm spatial at the plasma membrane of living host CD4+ T cells.
resolution and millisecond temporal resolution. HIV-1 particle assembly The modality of assembly for the Nipah virus was for the first time
at the host cell plasma membrane occurs via thousands of viral Gag pro- revealed by using an SMSM similar to a dSTORM combined with TIRF,
teins forming particles with a diameter of 100–130 nm. By introducing showing the virus assembly model previously undetermined using bio-
into the HIV-1 Gag precursor and derivatives, a photoactive fluorescent chemical analyses and electron microscopy [57].

9
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

Depth imaging Discussion and conclusions

Another relevant criterion for FSR methods to be applied to biologi- FSR methods have largely progressed from the time of their incep-
cal samples imaging and virus imaging and tracking is the sample depth tion, at a current stage of development and deployment which has pro-
penetration of the excitation beam. vided many commercial systems, that are progressively becoming of
Refractive index mismatch between air and tissue is responsible for common use in biomedical imaging laboratories. However, for virus
optical aberrations and light scattering, limiting factors to achieve res- imaging, there is not yet one perfect approach, as the above-described
olution in sample depth by using fluorescence microscopy. The limited techniques have a unique set of advantages and disadvantages. These
sample depth penetration is due to poor image resolution and limited approaches are evolving to combine more than one imaging method
contrast due to increased noise levels due to scattering. These effects to mitigate disadvantages and leverage on the respective advantages or
can be mitigated by using two-photon-based excitation at a longer laser simplicity of use. Current virology studies employing FSR imaging have
wavelength which reduces scattering [113], by the use of objective contributed to many novel insights even if still achieved mostly in fixed
lenses with fluid better matching the sample’s refractive index such as cells environment and in vitro cell cultures. As such their potentials to
glycerol-immersion microscope [114], or by the use of adaptive optics study the dynamic behavior of viruses’ components and their interac-
which can limit effects of optical aberrations [115]. tions with living cells or tissues environment, is not yet accomplished.
Light-sheet fluorescence microscopy (LSDM) [116] is another New development of FSR methods has recently seen the combination of
method that permits superior sample depth penetration allowing 3D STED and STORM (MINFLUX), merging the advantages of both meth-
imaging, while it does not improve spatial resolution, and it is advanta- ods, thus permitting to discern molecules with 1 nm precision at 9 nm
geous to combine it with FSR approaches for better 3D imaging of live distance and 100 times better temporal resolution in single molecules
cells [117,118]. This approach has been used for virus imaging com- tracking. Due to MINFLUX recent application in 3D imaging in fixed
bined with SIM and RESOLFT for herpesvirus [45,119]. The method and live cells [122], it could be further applied in observing the virus
permitted to image and track the herpesvirus capsids transport occur- components dynamics in living cells [123].
ring by diffusion. However, one of the major problems for expanding the use of FSR
LSDM is also used successfully for Single Virus tracking, together imaging in live-cells virology is the need for biosafety containment level
with other approaches described above when SR is needed. 3 or 4 (BSL-3 and BSL-4 laboratories), which are not very common. In
this regard, the development of commercial nano imagers could mit-
igate safety requirements as well the use of pseudovirus that can be
Single virus or particle tracking handled in biosafety level 2 laboratories in place of wild‐type viruses.
Pseudovirus [124], deprived of the certain virulent genome, can only
Single virus or particle tracking (SVT) is another method to image replicate once, and are obtained by combining proteins from the surfaces
viruses [7]. While FSR microscopy can provide nanometric resolution of other viruses, as an example for SARS-COV-2 virus with replication-
in imaging viruses, their trajectory tracking can be achieved without defective HIV or another retrovirus [125], could advance the in vivo
the need for FSR methods by simply using wide-field microscopy, TIRF FSR microscopy application to a virus.
microscopy, confocal microscopy, and spectroscopy. These methods al- Based on the so far few demonstrations and on the HIV-1 viruses
low for the imaging of virions labeled with photo-stable organic fluores- studies, where the key mechanisms of the viruses and the cell interaction
cent dyes or fluorescent proteins and permit tracking their position with and internalization were unveiled, the current methods can be applied to
highly accurate particle localization in the nanometer range better than other viruses for example SARS-CoV2, towards the development of drugs
the resolution of microscopes images [7]. For example in ref. [46], Ebola that specifically prevent these processes. There is in fact a clear need
virus dynamic entry and internalization into live cells were visualized to complement existing viral imaging techniques with super-resolution
by fluorescent virus labeling using SVT at the single virus level. This for methods for a better understanding of the SARS-CoV2 replication cycle
the first time permitted to observe that Ebola virus internalization in in presence of chemotherapeutic and chemopreventive agents [126].
cells occurs through lipid rafts. It can be finally concluded that the above-mentioned techniques
A label-free method has been used to track HIV such as interfero- could be implemented not only to advance virions’ imaging and mo-
metric scattering microscopy (iSCAT) [120]. iSCAT permits visualiza- tion analyses to provide information about biological mechanisms, as it
tion of the position of the virus using the interference of light owing to has been shown now in some examples, but also to facilitate the devel-
reflection from the surface of the coverslip and from the virus itself with opment of antiviral therapeutics and vaccines. This seems to be possible
high temporal and spatial resolution virus tracking. iSCAT in conjunc- at the current stage of development and even better when their full po-
tion with SMSM was used to investigate the diffusion of single Simian tential could be reached, which is in vivo imaging and single-particle
Virus 40 particles labeled with QDs. Due to the achieved 8 ms temporal tracking could be implemented in cultured cells with different thera-
resolution and 9 nm spatial resolution, it was possible to determine the peutics.
orientation of the virus on supported lipid bilayers [68].
Wide-field microscopy allows SVT in a large volume due to the large
depth of focus associated with a low signal loss in depth. It is better Declaration of Competing Interest
suited when there are low auto-fluorescence and low concentration and
distribution of fluorescent viruses. It has been used to study the trans- The authors declare no conflict of interest.
port, acidification, and fusion of single influenza viruses in living cells
and it was possible to separate distinct phases of the viral internaliza-
tion within cells [101]. However, wide-field microscopy with samples Acknowledgments
exhibiting a high background results in out-of-focus probes with low-
contrast images, thus lower resolution as it was revealed in HIV assem- The authors received no funding.
bly tracking by using fluorescence-labeled Gag protein [121].
SVT has been combined with FSR microscopy to increase the tempo-
ral resolution of PALM and STORM. Combining Single-particle tracking Supplementary materials
with PALM (known as SPT-PALM) enabled monitoring of the molecular
diffusion of single-molecule Gag and tsO45 proteins from the vesicular Supplementary material associated with this article can be found, in
stomatitis virus G (VSVG) [41]. the online version, at doi:10.1016/j.chphi.2021.100013.

10
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

References [34] S.W. Hell, S. Jakobs, L. Kastrup, Imaging and writing at the nanoscale with fo-
cused visible light through saturable optical transitions, Appl. Phys. A 77 (2003)
[1] S. Banerjee, S. Maurya, R. Roy, Single-molecule fluorescence imaging: Generating 859–860.
insights into molecular interactions in virology, 43 (2018) 519-540. [35] T. Grotjohann, I. Testa, M. Leutenegger, H. Bock, N.T. Urban, F. Lavoie-Cardinal,
[2] S. Rocha, J. Hendrix, D. Borrenberghs, Z. Debyser, J. Hofkens, Imaging the Repli- K.I. Willig, C. Eggeling, S. Jakobs, S.W. Hell, Diffraction-unlimited all-optical imag-
cation of Single Viruses: Lessons Learned from HIV and Future Challenges To Over- ing and writing with a photochromic GFP, Nature 478 (2011) 204–208.
come, ACS Nano 14 (2020) 10775–10783. [36] M. Hofmann, C. Eggeling, S. Jakobs, S.W. Hell, Breaking the diffraction barrier in
[3] I. Banerjee, Y. Yamauchi, A. Helenius, P. Horvath, High-content analysis of sequen- fluorescence microscopy at low light intensities by using reversibly photoswitch-
tial events during the early phase of influenza A virus infection, 8 (2013) e68450. able proteins, in: Proc. Natl. Acad. Sci. USA, 102, 2005, p. 17565.
[4] J.M. Coffin, S.H. Hughes, H.E. Varmus, The interactions of retroviruses and their [37] J. Grove, Super-resolution microscopy: a virus’ eye view of the cell, 6 (2014) 1365-
hosts, (1997). 1378.
[5] J. Shang, Y. Wan, C. Luo, G. Ye, Q. Geng, A. Auerbach, F. Li, Cell entry mechanisms [38] B. Müller, M. Heilemann, Shedding new light on viruses: Super-resolution mi-
of SARS-CoV-2, Proc. Natl. Acad. Sci. 117 (2020) 11727–11734. croscopy for studying human immunodeficiency virus, Trends Microbiol, 21
[6] I.H. Wang, C.J. Burckhardt, A. Yakimovich, U.F. Greber, Imaging, tracking and (2013).
computational analyses of virus entry and egress with the cytoskeleton, Viruses 10 [39] J. Hanne, V. Zila, M. Heilemann, B. Müller, H.-G. Kräusslich, Super-resolved in-
(2018) 166. sights into human immunodeficiency virus biology, 590 (2016) 1858-1876.
[7] S.-L. Liu, Z.-G. Wang, H.-Y. Xie, A.-A. Liu, D.C. Lamb, D.-W. Pang, Single-virus [40] K.I. Willig, R.R. Kellner, R. Medda, B. Hein, S. Jakobs, S.W. Hell, Nanoscale reso-
tracking: from imaging methodologies to virological applications, Chem. Rev. 120 lution in GFP-based microscopy, Nat. Methods 3 (2006) 721–723.
(2020) 1936–1979. [41] S. Manley, J.M. Gillette, G.H. Patterson, H. Shroff, H.F. Hess, E. Betzig, J. Lippin-
[8] N.d. Jonge, D.B. Peckys, G.J. Kremers, D.W. Piston, Electron microscopy of whole cott-Schwartz, High-density mapping of single-molecule trajectories with photoac-
cells in liquid with nanometer resolution, Proc. Natl. Acad. Sci. 106 (2009) 2159. tivated localization microscopy, Nat. Methods 5 (2008) 155–157.
[9] K.R. Richert-Pöggeler, K. Franzke, K. Hipp, R.G. Kleespies, Electron microscopy [42] D. Kim, T. Deerinck, Y. Sigal, H. Babcock, M. Ellisman, X. Zhuang, Correlative
methods for virus diagnosis and high resolution analysis of viruses, Front. Micro- stochastic optical reconstruction microscopy and electron microscopy, PLOS ONE
biol. 9 (2019). 10 (2015) e0124581.
[10] Y. Pan, S. Wang, Y. Shan, D. Zhang, J. Gao, M. Zhang, S. Liu, M. Cai, H. Xu, G. Li, [43] C. Qin, W. Li, Q. Li, W. Yin, X. Zhang, Z. Zhang, X.-E. Zhang, Z. Cui, Real-time
Q. Qin, H. Wang, Ultrafast Tracking of a Single Live Virion During the Invagination dissection of dynamic uncoating of individual influenza viruses, Proc. Natl. Acad.
of a Cell Membrane, Small 11 (2015) 2782–2788. Sci. 116 (2019) 2577–2582.
[11] A. Herrmann, C. Sieben, Single-virus force spectroscopy unravels molecular details [44] A. Kolpe, M. Arista-Romero, B. Schepens, S. Pujals, X. Saelens, L. Albertazzi, Su-
of virus infection, Integr. Biol. 7 (2015) 620–632. per-resolution microscopy reveals significant impact of M2e-specific monoclonal
[12] N. Parveen, D. Borrenberghs, S. Rocha, J. Hendrix, Single Viruses on the Fluores- antibodies on influenza A virus filament formation at the host cell surface, Sci.
cence Microscope: Imaging Molecular Mobility, Interactions and Structure Sheds Rep. 9 (2019) 4450.
New Light on Viral Replication, Viruses 10 (2018) 250. [45] J.B. Bosse, I.B. Hogue, M. Feric, S.Y. Thiberge, B. Sodeik, C.P. Brangwynne, L.W. En-
[13] E. Abbe, Beiträge zur Theorie des Mikroskops und der mikroskopischen quist, Remodeling nuclear architecture allows efficient transport of herpesvirus
Wahrnehmung, Archiv für mikroskopische Anatomie 9 (1873) 413–468. capsids by diffusion, Proc. Natl. Acad. Sci. 112 (2015) E5725.
[14] J. Chojnacki, C. Eggeling, Super-resolution fluorescence microscopy studies of hu- [46] C. Jin, B. Che, Z. Guo, C. Li, Y. Liu, W. Wu, S. Wang, D. Li, Z. Cui, M. Liang, Single
man immunodeficiency virus, Retrovirology 15 (2018) 41. virus tracking of Ebola virus entry through lipid rafts in living host cells, Biosaf.
[15] R.P.J. Nieuwenhuizen, K.A. Lidke, M. Bates, D.L. Puig, D. Grünwald, S. Stallinga, Health 2 (2020) 25–31.
B. Rieger, Measuring image resolution in optical nanoscopy, Nat. Methods 10 [47] J. Chojnacki, T. Staudt, B. Glass, P. Bingen, J. Engelhardt, M. Anders, J. Schneider,
(2013) 557–562. B. Müller, S.W. Hell, H.-G. Kräusslich, Maturation-dependent HIV-1 surface protein
[16] M.G.L. Gustafsson, Surpassing the lateral resolution limit by a factor of two using redistribution revealed by fluorescence nanoscopy, Science 338 (2012) 524.
structured illumination microscopy, J. Microsc. 198 (2000) 82–87. [48] J. Hanne, F. Göttfert, J.í. Schimer, M. Anders-Össwein, J. Konvalinka, J. En-
[17] M.G.L. Gustafsson, Nonlinear structured-illumination microscopy: Wide-field flu- gelhardt, B. Müller, S.W. Hell, H.-G. Kräusslich, Stimulated emission depletion
orescence imaging with theoretically unlimited resolution, Proc. Natl. Acad. Sci. nanoscopy reveals time-course of human immunodeficiency virus proteolytic mat-
USA 102 (2005) 13081. uration, ACS nano 10 (2016) 8215–8222.
[18] B. Huang, H. Babcock, X. Zhuang, Breaking the diffraction barrier: super-resolution [49] A. Honigmann, V. Mueller, H. Ta, A. Schoenle, E. Sezgin, S.W. Hell, C. Eggeling,
imaging of cells, Cell 143 (2010) 1047–1058. Scanning STED-FCS reveals spatiotemporal heterogeneity of lipid interaction in the
[19] E. Betzig, G.H. Patterson, R. Sougrat, O.W. Lindwasser, S. Olenych, J.S. Bonifacino, plasma membrane of living cells, Nat. Commun. 5 (2014) 5412.
M.W. Davidson, J. Lippincott-Schwartz, H.F. Hess, imaging intracellular fluorescent [50] C.F. Pereira, J. Rossy, D.M. Owen, J. Mak, K. Gaus, HIV taken by STORM: Super-res-
proteins at nanometer resolution, Science 313 (2006) 1642. olution fluorescence microscopy of a viral infection, Virol. J. 9 (2012) 84.
[20] S.T. Hess, T.P. Girirajan, M.D. Mason, Ultra-high resolution imaging by fluores- [51] M. Lehmann, S. Rocha, B. Mangeat, F. Blanchet, H. Uji-i, J. Hofkens, V. Piguet,
cence photoactivation localization microscopy, Biophys. J. 91 (2006) 4258–4272. Quantitative multicolor super-resolution microscopy reveals tetherin HIV-1 inter-
[21] S.W. Hell, Far-field optical nanoscopy, Science 316 (2007) 1153–1158. action, PLOS Pathogens 7 (2011) e1002456.
[22] M.J. Rust, M. Bates, X. Zhuang, Sub-diffraction-limit imaging by stochastic optical [52] W. Muranyi, S. Malkusch, B. Müller, M. Heilemann, H.-G. Kräusslich, Super-reso-
reconstruction microscopy (STORM), Nat. Methods 3 (2006) 793–796. lution microscopy reveals specific recruitment of HIV-1 envelope proteins to viral
[23] M. Heilemann, S. van de Linde, M. Schüttpelz, R. Kasper, B. Seefeldt, A. Mukher- assembly sites dependent on the envelope C-terminal tail, PLOS Pathogens 9 (2013)
jee, P. Tinnefeld, M. Sauer, Subdiffraction-resolution fluorescence imaging with e1003198.
conventional fluorescent probes, Angew. Chem. Int. Ed. 47 (2008) 6172–6176. [53] Y.-C. Chen, C. Sood, M. Marin, J. Aaron, E. Gratton, K. Salaita, G.B. Melikyan,
[24] S.W. Hell, J. Wichmann, Breaking the diffraction resolution limit by stimulated Super-resolution fluorescence imaging reveals that serine incorporator protein 5
emission: stimulated-emission-depletion fluorescence microscopy, Opt. Lett. 19 inhibits human immunodeficiency virus fusion by disrupting envelope glycoprotein
(1994) 780–782. clusters, ACS Nano 14 (2020) 10929–10943.
[25] J. Huff, The Airyscan detector from ZEISS: confocal imaging with improved signal– [54] C. Floderer, J.-B. Masson, E. Boilley, S. Georgeault, P. Merida, M. El Beheiry,
to-noise ratio and super-resolution, Nat. Methods 12 (2015) i–ii. M. Dahan, P. Roingeard, J.-B. Sibarita, C. Favard, Single molecule localisation mi-
[26] P. Monaghan, D. Green, J. Pallister, R. Klein, J. White, C. Williams, P. McMillan, croscopy reveals how HIV-1 Gag proteins sense membrane virus assembly sites in
L. Tilley, M. Lampe, P. Hawes, L.-F. Wang, Detailed morphological characterisa- living host CD4 T cells, Sci. Rep. 8 (2018) 1–15.
tion of Hendra virus infection of different cell types using super-resolution and [55] R.F. Laine, A. Albecka, S. van de Linde, E.J. Rees, C.M. Crump, C.F. Kaminski,
conventional imaging, Virol. J. 11 (2014) 200. Structural analysis of herpes simplex virus by optical super-resolution imaging,
[27] L.A. Caldas, R.C. Azevedo, J.L. da Silva, W. de Souza, Microscopy analysis of Zika Nat. Commun. 6 (2015) 5980.
virus morphogenesis in mammalian cells, Sci. Rep. 10 (2020) 8370. [56] F. Beilstein, G.H. Cohen, R.J. Eisenberg, V. Nicolas, A. Esclatine, D. Pasdeloup,
[28] J. Horsington, L. Turnbull, C.B. Whitchurch, T.P. Newsome, Sub-viral imaging of Dynamic organization of Herpesvirus glycoproteins on the viral envelope revealed
vaccinia virus using super-resolution microscopy, J. Virol. Methods 186 (2012) by super-resolution microscopy, PLOS Pathogens 15 (2019) e1008209.
132–136. [57] Q. Liu, L. Chen, H.C. Aguilar, K.C. Chou, A stochastic assembly model for Nipah
[29] K.N. Fish, Total internal reflection fluorescence (TIRF) microscopy, Curr. Protoc. virus revealed by super-resolution microscopy, Nat. Commun. 9 (2018) 3050.
Cytom. Chapter 12 (2009) Unit12.18. [58] S.-L. Liu, Z.-L. Zhang, Z.-Q. Tian, H.-S. Zhao, H. Liu, E.-Z. Sun, G.F. Xiao, W. Zhang,
[30] D. Li, L. Shao, B.-C. Chen, X. Zhang, M. Zhang, B. Moses, D.E. Milkie, J.R. Beach, H.-Z. Wang, D.-W. Pang, Effectively and efficiently dissecting the infection of in-
J.A. Hammer, M. Pasham, T. Kirchhausen, M.A. Baird, M.W. Davidson, P. Xu, fluenza virus by quantum-dot-based single-particle tracking, ACS Nano 6 (2012)
E. Betzig, Extended-resolution structured illumination imaging of endocytic and 141–150.
cytoskeletal dynamics, Science 349 (2015) aab3500. [59] F. Baharom, O.S. Thomas, R. Lepzien, I. Mellman, C. Chalouni, A. Smed-Sörensen,
[31] S. Cox, E. Rosten, J. Monypenny, T. Jovanovic-Talisman, D.T. Burnette, J. Lippin- Visualization of early influenza A virus trafficking in human dendritic cells using
cott-Schwartz, G.E. Jones, R. Heintzmann, Bayesian localization microscopy re- STED microscopy, PLOS ONE 12 (2017) e0177920.
veals nanoscale podosome dynamics, Nat. Methods 9 (2012) 195–200. [60] M. Mehedi, M. Smelkinson, J. Kabat, S. Ganesan, P.L. Collins, U.J. Buchholz, Mul-
[32] Y. Garcés, J. Martínez Guevara, D. Hernandez, H. Hernández, A. Pérez-Delgado, ticolor stimulated emission depletion (STED) microscopy to generate high-resolu-
M. Méndez, C. Wood, J. Rendon-Mancha, D. Silva-Ayala, S. López, A. Guerrero, tion images of respiratory syncytial virus particles and infected cells, Bio. Protoc.
C. Arias, Nanoscale organization of rotavirus replication machineries, eLife 8 7 (2017) e2543.
(2019). [61] B. Huang, M. Bates, X. Zhuang, Super-resolution fluorescence microscopy, Annu.
[33] B. Huang, W. Wang, M. Bates, X. Zhuang, Three-dimensional super-resolution imag- Rev. Biochem. 78 (2009) 993–1016.
ing by stochastic optical reconstruction microscopy, Science 319 (2008) 810–813.

11
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

[62] L. Schermelleh, A. Ferrand, T. Huser, C. Eggeling, M. Sauer, O. Biehlmaier, [92] A.A. Waheed, E.O. Freed, HIV type 1 Gag as a target for antiviral therapy, AIDS
G.P.C. Drummen, Super-resolution microscopy demystified, Nat. Cell Biol. 21 Res. Hum. Retroviruses 28 (2012) 54–75.
(2019) 72–84. [93] J. Hanne, F. Göttfert, J. Schimer, M. Anders-Össwein, J. Konvalinka, J. Engelhardt,
[63] D. Wojta-Stremayr, W.F. Pickl, Fluorosomes: fluorescent virus-like nanoparticles B. Müller, S.W. Hell, H.-G. Kräusslich, Stimulated emission depletion nanoscopy
that represent a convenient tool to visualize receptor-ligand interactions, 13 (2013) reveals time-course of human immunodeficiency virus proteolytic maturation, ACS
8722-8749. Nano 10 (2016) 8215–8222.
[64] D. Kamiyama, S. Sekine, B. Barsi-Rhyne, J. Hu, B. Chen, L.A. Gilbert, H. Ishikawa, [94] V. Sakin, J. Hanne, J. Dunder, M. Anders-Össwein, V. Laketa, I. Nikić, H.-G. Kräus-
M.D. Leonetti, W.F. Marshall, J.S. Weissman, B. Huang, Versatile protein tagging slich, E.A. Lemke, B.J.C.c.b. Müller, A versatile tool for live-cell imaging and super-
in cells with split fluorescent protein, Nat. Commun. 7 (2016) 11046. resolution nanoscopy studies of HIV-1 Env distribution and mobility, 24 (2017)
[65] S.W. Hell, S.J. Sahl, M. Bates, X. Zhuang, R. Heintzmann, M.J. Booth, J. Bewersdorf, 635-645. e635.
G. Shtengel, H. Hess, P. Tinnefeld, The 2015 super-resolution microscopy roadmap, [95] J. Chojnacki, D. Waithe, P. Carravilla, N. Huarte, S. Galiani, J. Enderlein,
J. Phys. D: Appl. Phys. 48 (2015) 443001. C. Eggeling, Envelope glycoprotein mobility on HIV-1 particles depends on the
[66] V. Sakin, G. Paci, E.A. Lemke, B. Müller, Labeling of virus components for advanced, virus maturation state, Nat. Commun. 8 (2017) 545.
quantitative imaging analyses, FEBS Lett. 590 (2016) 1896–1914. [96] P. Schwille, Fluorescence correlation spectroscopy and its potential for intracellular
[67] M. Heilemann, S. van de Linde, A. Mukherjee, M. Sauer, Super-resolution imaging applications, Cell Biochem. Biophys. 34 (2001) 383–408.
with small organic fluorophores, Angew. Chem. Int. Ed. 48 (2009) 6903–6908. [97] C. Eggeling, C. Ringemann, R. Medda, G. Schwarzmann, K. Sandhoff, S. Polyakova,
[68] P. Kukura, H. Ewers, C. Müller, A. Renn, A. Helenius, V. Sandoghdar, High-speed V.N. Belov, B. Hein, C. von Middendorff, A. Schönle, S.W. Hell, Direct observation
nanoscopic tracking of the position and orientation of a single virus, Nat. Methods of the nanoscale dynamics of membrane lipids in a living cell, Nature 457 (2009)
6 (2009) 923–927. 1159–1162.
[69] C. Lv, Y. Lin, A.-A. Liu, Z.-Y. Hong, L. Wen, Z. Zhang, Z.-L. Zhang, H. Wang, [98] K. Inamdar, C. Floderer, C. Favard, D. Muriaux, Monitoring HIV-1 assembly in
D.-W. Pang, Labeling viral envelope lipids with quantum dots by harnessing the living cells: insights from dynamic and single molecule microscopy, Viruses 11
biotinylated lipid-self-inserted cellular membrane, Biomaterials 106 (2016) 69–77. (2019) 72.
[70] S.-L. Liu, Z.-Q. Tian, Z.-L. Zhang, Q.-M. Wu, H.-S. Zhao, B. Ren, D.-W. Pang, High– [99] L.M. Molinos-Albert, B. Clotet, J. Blanco, J. Carrillo, Immunologic insights on
efficiency dual labeling of influenza virus for single-virus imaging, Biomaterials 33 the membrane proximal external region: a major human immunodeficiency virus
(2012) 7828–7833. type-1 vaccine target, Front. Immunol. 8 (2017) 1154.
[71] A. Baron, N. Osipov, S. Yashchenko, Y.A. Kokotukha, I. Baron, A. Puzyr, [100] P. Carravilla, J. Chojnacki, E. Rujas, S. Insausti, E. Largo, D. Waithe, B. Apellaniz,
I. Olkhovskiy, V. Bondar, Adsorption of viral particles from the blood plasma of T. Sicard, J.-P. Julien, C. Eggeling, J.L. Nieva, Molecular recognition of the native
patients with viral hepatitis on nanodiamonds, in: Doklady Biochemistry and Bio- HIV-1 MPER revealed by STED microscopy of single virions, Nat. Commun. 10
physics, Springer, 2016, pp. 244–246. (2019) 78.
[72] M. Gu, Y. Cao, S. Castelletto, B. Kouskousis, X. Li, Super-resolving single nitrogen [101] M. Lakadamyali, M.J. Rust, H.P. Babcock, X. Zhuang, Visualizing infection of indi-
vacancy centers within single nanodiamonds using a localization microscope, Opt. vidual influenza viruses, Proc. Natl. Acad. Sci. 100 (2003) 9280.
Express 21 (2013) 17639–17646. [102] T. Dertinger, R. Colyer, G. Iyer, S. Weiss, J. Enderlein, Fast, background-free, 3D su-
[73] M. Barbiero, S. Castelletto, X. Gan, M. Gu, Spin-manipulated nanoscopy for single per-resolution optical fluctuation imaging (SOFI), Proc. Natl. Acad. Sci. 106 (2009)
nitrogen-vacancy center localizations in nanodiamonds, Light: Sci. Appl. 6 (2017) 22287.
e17085 -e17085. [103] D. Baddeley, I.D. Jayasinghe, C. Cremer, M.B. Cannell, C. Soeller, Light-induced
[74] E.H. Chen, O. Gaathon, M.E. Trusheim, D. Englund, Wide-field multispectral su- dark states of organic fluochromes enable 30 nm resolution imaging in standard
per-resolution imaging using spin-dependent fluorescence in nanodiamonds, Nano media, Biophys. J. 96 (2009) L22–L24.
Lett. 13 (2013) 2073–2077. [104] E. Jensen, D.J. Crossman, Technical review: types of imaging—direct STORM,
[75] S. Arroyo-Camejo, M.-P. Adam, M. Besbes, J.-P. Hugonin, V. Jacques, J.-J. Greffet, Anatom. Rec. 297 (2014) 2227–2231.
J.-F. Roch, S.W. Hell, F. Treussart, Stimulated emission depletion microscopy re- [105] G.T. Dempsey, J.C. Vaughan, K.H. Chen, M. Bates, X. Zhuang, Evaluation of fluo-
solves individual nitrogen vacancy centers in diamond nanocrystals, ACS Nano 7 rophores for optimal performance in localization-based super-resolution imaging,
(2013) 10912–10919. Nat. Methods 8 (2011) 1027–1036.
[76] P. Maurer, J. Maze, P. Stanwix, L. Jian, A. Gorshkov, A. Zibrov, B. Harke, J. Hodges, [106] S. van de Linde, U. Endesfelder, A. Mukherjee, M. Schüttpelz, G. Wiebusch,
A. Zibrov, A. Yacoby, D. Twitchen, S. Hell, R. Walsworth, M. Lukin, Far-field optical S. Wolter, M. Heilemann, M. Sauer, Multicolor photoswitching microscopy for sub-
imaging and manipulation of individual spins with nanoscale resolution, Nat. Phys. diffraction-resolution fluorescence imaging, Photochem. Photobiol. Sci. 8 (2009)
6 (912-918) (2010) 6 (2010). 465–469.
[77] M. Barbiero, S. Castelletto, Q. Zhang, Y. Chen, M. Charnley, S. Russell, M. Gu, [107] H. Shroff, C.G. Galbraith, J.A. Galbraith, H. White, J. Gillette, S. Olenych,
Nanoscale magnetic imaging enabled by nitrogen vacancy centres in nanodiamonds M.W. Davidson, E. Betzig, Dual-color superresolution imaging of genetically ex-
labelled by iron–oxide nanoparticles, Nanoscale 12 (2020) 8847–8857. pressed probes within individual adhesion complexes, Proc. Natl. Acad. Sci. 104
[78] M. Pfender, N. Aslam, G. Waldherr, P. Neumann, J. Wrachtrup, Single-spin stochas- (2007) 20308.
tic optical reconstruction microscopy, Proc. Natl. Acad. Sci. 111 (2014) 14669. [108] H. Bock, C. Geisler, C.A. Wurm, C. Von Middendorff, S. Jakobs, A. Schönle, A. Eg-
[79] G.E. Johnstone, G.S. Cairns, B.R. Patton, Nanodiamonds enable adaptive-optics en- ner, S.W. Hell, C. Eggeling, Two-color far-field fluorescence nanoscopy based on
hanced, super-resolution, two-photon excitation microscopy, R. Soc. Open Sci. 6 photoswitchable emitters, Appl. Phys. B 88 (2007) 161–165.
(2019) 190589, doi:10.1098/rsos.190589. [109] M. Bates, B. Huang, G.T. Dempsey, X. Zhuang, Multicolor super-resolution imaging
[80] E. Rittweger, K.Y. Han, S.E. Irvine, C. Eggeling, S.W. Hell, STED microscopy reveals with photo-switchable fluorescent probes, Science 317 (2007) 1749–1753.
crystal colour centres with nanometric resolution, Nat. Photon. 3 (2009) 144. [110] M.F. Juette, T.J. Gould, M.D. Lessard, M.J. Mlodzianoski, B.S. Nagpure, B.T. Ben-
[81] J.-I. Chao, E. Perevedentseva, P.-H. Chung, K.-K. Liu, C.-Y. Cheng, C.-C. Chang, nett, S.T. Hess, J. Bewersdorf, Three-dimensional sub–100 nm resolution fluores-
C.-L. Cheng, Nanometer-sized diamond particle as a probe for biolabeling, Biophys. cence microscopy of thick samples, Nat. Methods 5 (2008) 527–529.
J. 93 (2007) 2199–2208. [111] S.A. Jones, S.-H. Shim, J. He, X. Zhuang, Fast, three-dimensional super-resolution
[82] S. Castelletto, M. Barbiero, M. Charnley, A. Boretti, M. Gu, Imaging with nanometer imaging of live cells, Nat. Methods 8 (2011) 499–505.
resolution using optically active defects in silicon carbide, Phys. Rev. Appl. 14 [112] N. Gustafsson, S. Culley, G. Ashdown, D.M. Owen, P.M. Pereira, R. Henriques, Fast
(2020) 034021. live-cell conventional fluorophore nanoscopy with ImageJ through super-resolu-
[83] J. Feng, H. Deschout, S. Caneva, S. Hofmann, I. Lončarić, P. Lazić, A. Radenovic, tion radial fluctuations, Nat. Commun. 7 (2016) 12471.
Imaging of optically active defects with nanometer resolution, Nano Lett. 18 (2018) [113] K.T. Takasaki, J.B. Ding, B.L. Sabatini, Live-cell superresolution imaging by pulsed
1739–1744. STED two-photon excitation microscopy, Biophys. J. 104 (2013) 770–777.
[84] C. Eggeling, K.I. Willig, S.J. Sahl, S.W. Hell, Lens-based fluorescence nanoscopy, [114] N.T. Urban, K.I. Willig, S.W. Hell, U.V. Nägerl, STED nanoscopy of actin dynamics
Q. Rev. Biophys. 48 (2015) 178–243. in synapses deep inside living brain slices, Biophys. J. 101 (2011) 1277–1284.
[85] B. Harke, J. Keller, C.K. Ullal, V. Westphal, A. Schönle, S.W. Hell, Resolution scaling [115] T.J. Gould, D. Burke, J. Bewersdorf, M.J. Booth, Adaptive optics enables 3D STED
in STED microscopy, Opt. Express 16 (2008) 4154–4162. microscopy in aberrating specimens, Opt. Express 20 (2012) 20998–21009.
[86] V. Westphal, S.W. Hell, Nanoscale resolution in the focal plane of an optical mi- [116] E.H.K. Stelzer, Light-sheet fluorescence microscopy for quantitative biology, Nat.
croscope, Phys. Rev. Lett. 94 (2005) 143903. Methods 12 (2015) 23–26.
[87] R. Schmidt, C.A. Wurm, S. Jakobs, J. Engelhardt, A. Egner, S.W. Hell, Spherical [117] F. Cella Zanacchi, Z. Lavagnino, M. Perrone Donnorso, A. Del Bue, L. Furia,
nanosized focal spot unravels the interior of cells, Nat. Methods 5 (2008) 539–544. M. Faretta, A. Diaspro, Live-cell 3D super-resolution imaging in thick biological
[88] U.V. Nägerl, K.I. Willig, B. Hein, S.W. Hell, T. Bonhoeffer, Live-cell imaging of samples, Nat. Methods 8 (2011) 1047–1049.
dendritic spines by STED microscopy, Proc. Natl. Acad. Sci. 105 (2008) 18982. [118] L. Gao, L. Shao, C. Higgins, J. Poulton, M. Peifer, M. Davidson, X. Wu, B. Goldstein,
[89] F. Bottanelli, E.B. Kromann, E.S. Allgeyer, R.S. Erdmann, S.Wood Baguley, G. Siri- E. Betzig, Noninvasive Imaging beyond the Diffraction Limit of 3D Dynamics in
nakis, A. Schepartz, D. Baddeley, D.K. Toomre, J.E. Rothman, J. Bewersdorf, Two– Thickly Fluorescent Specimens, Cell 151 (2012) 1370–1385.
colour live-cell nanoscale imaging of intracellular targets, Nat. Commun. 7 (2016) [119] P. Hoyer, G. de Medeiros, B. Balázs, N. Norlin, C. Besir, J. Hanne, H.-G. Kräusslich,
10778. J. Engelhardt, S.J. Sahl, S.W. Hell, L. Hufnagel, Breaking the diffraction limit of
[90] V. Westphal, S.O. Rizzoli, M.A. Lauterbach, D. Kamin, R. Jahn, S.W. Hell, light-sheet fluorescence microscopy by RESOLFT, Proc. Natl. Acad. Sci. 113 (2016)
Video-rate far-field optical nanoscopy dissects synaptic vesicle movement, Science 3442.
320 (2008) 246. [120] J. Ortega-Arroyo, P. Kukura, Interferometric scattering microscopy (iSCAT): new
[91] J. Schneider, J. Zahn, M. Maglione, S.J. Sigrist, J. Marquard, J. Chojnacki, frontiers in ultrafast and ultrasensitive optical microscopy, Phys. Chem. Chem.
H.-G. Kräusslich, S.J. Sahl, J. Engelhardt, S.W. Hell, Ultrafast, temporally stochastic Phys. 14 (2012) 15625–15636.
STED nanoscopy of millisecond dynamics, Nat. Methods 12 (2015) 827–830.

12
S. Castelletto and A. Boretti Chemical Physics Impact 2 (2021) 100013

[121] S. Ivanchenko, W.J. Godinez, M. Lampe, H.-G. Kräusslich, R. Eils, K. Rohr, [124] Q. Li, Q. Liu, W. Huang, X. Li, Y. Wang, Current status on the development of
C. Bräuchle, B. Müller, D.C. Lamb, Dynamics of HIV-1 assembly and release, PLOS pseudoviruses for enveloped viruses, Rev. Med. Virol. 28 (2018) e1963.
Pathogens 5 (2009) e1000652. [125] S.-W. Huang, C.-H. Tai, Y.-M. Hsu, D. Cheng, S.-J. Hung, K.M. Chai, Y.-F. Wang,
[122] K.C. Gwosch, J.K. Pape, F. Balzarotti, P. Hoess, J. Ellenberg, J. Ries, S.W. Hell, J.-R. Wang, Assessing the application of a pseudovirus system for emerging SARS–
MINFLUX nanoscopy delivers 3D multicolor nanometer resolution in cells, Nat. CoV-2 and re-emerging avian influenza virus H5 subtypes in vaccine development,
Methods 17 (2020) 217–224. Biomed. J. (2020).
[123] F. Balzarotti, Y. Eilers, K. Gwosch, A. Gynnå, V. Westphal, F. Stefani, J. Elf, S. Hell, [126] A. Boretti, B. Banik, S. Castelletto, Mechanism of action of Chloro-
Nanometer resolution imaging and tracking of fluorescent molecules with minimal quine/Hydroxychloroquine for Covid19 infection, Coronaviruses (2020).
photon fluxes, Science (2016) 355.

13

You might also like