Live Cell Imaging
Jennifer Walker-Daniels1 (jlwd at mail dot iastate dot edu), Orestis Faklaris2 (oresfak at gmail dot com)
1 Iowa State University, United States. 2 Centre National de la Recherche Scientifique, Montpellier, France
DOI
//dx.doi.org/10.13070/mm.en.2.124
Date
last modified : 2022-10-20; original version : 2012-07-23
Cite as
MATER METHODS 2012;2:124
Abstract

An overview of live cell imaging methods.

Introduction

Live cell imaging has revolutionized how biologists study cells, proteins and a multitude of processes and molecular interactions. Live cell imaging techniques allow scientists to observe cell structures and processes in real time, and over time. The observation of dynamic changes provides more insight into the operations of a cell than a snapshot provided by imaging fixed cells. Moreover, the spatial resolution of light microscopy allows observing subcellular structures, by using a range of fluorescent probes. Besides, since live cell imaging is less prone to experimental artifacts, it usually provides more reliable and relevant information than fixed cell microscopy. This review examines live cell imaging systems, fluorescent probes, methodologies, and applications. Jost AP and Waters JC published an excellent article about how to prevent errors in using microscopes, especially when quantitative experiments are performed [6].

Intravital microscopy observes physiological and cellular processes in live animals (for example, observing macrophages and endosomes inside mouse peritoneal serosa [7] ) and is not discussed here.

Applications of Live Cell Imaging

A wide array of biological questions exist that can be answered with live cell imaging. A few of the most popular applications include the examination of structural components of a cell, the study of dynamic processes, for example, chromatin looping [8], and the localization of molecules. Cellular integrity, endocytosis, exocytosis, protein trafficking, signal transduction, and enzyme activity can all be monitored. It is possible to study how molecules translocate, interact and respond to environmental cues. Furthermore, some imaging systems are capable of monitoring molecules within live animals. A recent example is the tracing of neural stem and progenitor cells in adult mouse brains for up to 2 months [9]. Table 1 shows the most common live-cell microscopy approaches and their current biological applications.

Biological application Microscopy Approach Abbreviation
Cell shape – cell migration – organelle kineticsTransmission Microscopy : Bright Field, Dark Field, Phase Contrast, Differential Interference ContrastBF / PH / DIC
Protein colocalization - dynamics, organisation and structure of cells , organelles, proteinsEpifluorescence / Confocal / Spinning disk
3D imaging of live cellsConfocal / Spinning disk
3D imaging of tissues, model organisms, small animals – study structure and developmentMultiphoton microscopy / light sheet microscopy
Quantification of ion concentrations (calcium, magnesium)ion imaging
Biosensing and protein-protein interactionsForster resonance energy transferFRET
Biosensing and protein-protein interactions without light excitationBioluminescence resonance energy transferBRET
Protein diffusion and kineticsFluorescence recovery after photo-bleachingFRAP
Protein diffusion with high spatio-temporal accuracy (μs-s and 300-400nm)Fluorescence correlation spectroscopyFCS
Protein diffusion with single molecule mapping and high spatio-temporal resolutionSingle particle trackingSPT
Protein diffusion with single molecule mapping and high spatio-temporal resolution in dense samplesSPT photoactivated localisation microscopysptPALM
Table 1. Live cell microscopy approaches utilized for common biological applications and their abbreviations.
Maintenance of Cell Health

The first concern for live cell imaging is to provide to cells an optimal physiological environment. In doing so, the scientist ensures that the cells not only stay alive but remain in a metabolic state with no nonspecific changes that could alter the process being observed. For short term imaging needs, cell health is less critical, but when imaging over hours or days, it is imperative to address the causes of altered metabolic function. The approaches include controlling pH, oxygenation, temperature, and osmolarity [10]. To maintain a physiological pH one has to control the CO2 concentration of the medium. Cells grow well in medium containing 10% fetal bovine serum and 5% CO2 environment. Special incubation chambers can be used that provide 5% CO2 concentration thanks to a pressure regulator. Another less expensive but often less efficient for long-term studies (>12h) solution is the HEPES buffer. The medium is buffered at pH 7.4 with HEPES and cells proliferate and can be kept alive at least for short-term experiments. Oxygenation is maintained for longer-term experiments by either changing the media often or using a larger volume of media initially. Osmolarity is kept near 300mosM and is maintained by the prevention of evaporation via a sealed chamber or the use of a humidified environment. To ensure that the air is humidified, the CO2 gas is bubbled through a water reservoir. The maintenance of optimal cell temperature is critical because fluctuations of just a few degrees can disrupt cellular physiology [10]. Temperature can be controlled by either using stage-top incubation chambers to heat the sample or large incubators locally to heat the whole microscope [11]. The stage top chambers fit in the XY stage or piezo insert of the microscope. They are connected to the Temperature and CO2 controller and create the appropriate environment for imaging health live cells. The objective should also be heated because it can act as a heat sink. Objective heater options include a metal foil blanket, copper tubing water jackets, and closed loop heaters. Temperature is also maintained by having an adequate volume of media in the cell chamber which serves as a thermal mass to reduce temperature fluctuations.

For better long-term temperature stability large boxes that enclose the specimen, stage, objective and part or the whole microscope body are preferred. They offer a better thermal equilibration, prevent temperature gradients and keep all parts of the scope at a common temperature, something that reduces mechanical drifts of the stage during acquisition. They often need to be custom-designed to fit around the microscope components. If at a later time new components are added to the microscope (e.g., cameras) the boxes should be adapted appropriately. They often take a long time to stabilize (several hours), they are more expensive and sometimes they can limit the direct access to all microscope components.

Imaging chambers

The cell chamber during imaging must provide conditions that keep the cells functioning and allow observation with the microscope objective. Most often live cells are viewed with an inverted microscope and are grown on glass coverslips. Like for fixed cell imaging, most of the coverslips should be type No 1.5 (0.17 mm thickness), which is the thickness for which most objectives are corrected to minimize spherical aberrations.

Various types of imaging chambers exist, from the simplest sealed coverslip on a microscope slide to perfusion chambers that enable control of the environment variables. They should all allow observation of the living samples with minimal invasion. They should be easily sterilized and isolated from the environment with a cover or seal during the observation, to avoid contamination. Sometimes it is also required that they offer access to the cells for the addition of reagents, changes of the medium or microinjections.

For short term imaging experiments (30 minutes or less), cells can perform well with just a coverslip over a spacer that is sealed with a sealant to prevent drying. Other coverslips or thin rubber slices are often used as spacers for this type of cell chamber. Sealant options include molten agarose, vacuum grease, or VALAP (1:1:1 mixture of Vaseline, lanolin, paraffin) which eliminate evaporation and restrict gas exchange.

Cell Health ParameterShort-Term StudyLonger-Term Study
pHHEPES BufferCO2 Incubator
TemperatureStage WarmersObjective Lens Warmers
Cell chambers with integrated heating elements
Enclose microscope within a heated box
HumidityOpen cell chambersTightly sealed cell chambers
OxygenationLarge volume of mediaMedia changes during study
Large media volume
OsmolaritySealed chamberEnclose system within a humidified chamber
Table 2. Summary of the how cell health is maintained during short or longer term live cell imaging studies.

For longer-term imaging, cell chambers with more advanced features are necessary. Open cell chambers are similar to Petri dishes in that they provide access to the atmosphere. These chambers allow access to the cells and thereby allow the researcher to add drugs, change the media, or perform microinjections. Closed cell chambers are sealed to prevent evaporation of the culture media and to insulate the cells from the outside environment. These may include a port that allows the addition of drug treatment. Simple sealed cell chambers include tissue culture plates that have a hole in the bottom where a coverslip may be placed, or microscope slides with small chambers sealed into the glass. Several commercial solutions exist either in a 35 mm round dish (MatTek) [12] or in a rectangular format (LabTek) [13] or in a multiple well format such as glass bottom chamber slides from ibidi (80286) [14] or Automation 96-well glass bottom black plates from Brooks [15]. Greenwood DJ et al, for example, seeded differentiated macrophages on gridded MatTek dishes (P35G-2-14-CGRD) for live-cell imaging [16]. Glass bottom dishes can also be self-made by drilling a hole in the plastic culture dish and gluing the glass cover slide on the bottom.

These chambers are simple to use but are not tightly sealed. One way to seal is spreading some silicon grease around the inner edge of the lid before closing the dish. An alternative is to spread a layer of mineral oil on the top of the culture and observation medium.

For cell-imaging experiments that require more precise environmental control, sealed closed chambers are recommended. The choice of the imaging chamber then is strongly related to the specimen and the duration of the experiment and should be coupled with the appropriate heating system of the specimen.

Commonly used chambers include 35 mm imaging µ-dishes with polymer coverslip bottom from Ibidi [17, 18].

The microscope and the acquisition

System attributes that provide optimal imaging conditions without affecting a lot the cell health are essential for live cell imaging. The filters of the excitation and emission light should be optimized to match the fluorophore, excite efficiently and detect the maximum of the emitted light. The use of fast mechanical shutters is essential for controlling the excitation light. For faster solutions, the shutters are triggered by the hardware (TTL trigger by the camera for example). Nowadays some excitation sources (LEDs and some lasers) can be switched on and off rapidly without the use of shutters.

For live imaging, motorized stages are recommended, that allow parallel acquisition of multiple regions of the specimen. Due to temperature gradients and stage stability, mechanical drift can appear during the experiments. To limit the focus drift, there are focus correction strategies, based either on software solutions (named autofocus) or on hardware, like the reflection of a laser on the cover slide. The latter is more efficient and should be preferred for the long term and multi-position acquisitions.

The detection system must be sensitive enough to obtain images from weakly fluorescent samples while acquiring a high signal level above any background noise or auto-fluorescence present in the sample. Cooled detectors are preferred to limit the readout noise. The detector must be fast enough to capture dynamic processes. The camera must possess enough resolution capacity to allow the capture of very fine details. The detection system must also provide a wide dynamic range to measure tiny differences in intensity accurately. The choice of the detector depends strongly on the needs of the experiment. For instance, for live cells with weak signals, EM-CCD is commonly used (back-thinned EM-CCD sensors are characterized by a high quantum efficiency that can be close to 95%). However, for fast live imaging, with a high spatial resolution, sCMOS cameras are preferred, because most often the pixel size of the chip of an sCMOS camera is around 6.5 μm when for EM-CCD the pixel size is commonly 16 μm. Significant progress is being carried out on sCMOS detectors, either by increasing their quantum efficiency with back-illuminated solutions or by lowering their noise level to crease the S/N ratio finally.

When performing live cell imaging, one should pay great attention to the illumination quantity that cells will receive and try always to minimize it to keep cells healthy and avoid phototoxicity [19]. Most cells and tissues do not usually encounter light exposure, and UV light is known to cause DNA damage. Following the excitation of fluorescent proteins or dye molecules, free radicals are formed and interact with surrounding cellular components. Efficient microscope design can minimize the detrimental effects of illumination. Microscopes should reduce the amount of excitation light used by having an optimal light path through the scope, and they should also utilize detectors optimized to detect the majority of the fluorescence emission. Within the parameters of individual experiments, illumination can be minimized by using the lowest concentration of a fluorescent probe that is needed to detect a signal. Phenol red-free medium can also help by reducing the background and exciting less (phenol red has a high visible light absorption extinction coefficient).

However, implementation of all of these techniques to minimize illumination is still not enough to completely prevent all light-induced disruptions, so controls should be performed with every experiment to ensure that cellular function has not been adversely affected by the imaging process.

Labeling of Intracellular Moieties
Fluorescent tags

The use of fluorescent probes has tremendously advanced the observation of cellular structures and processes. An ideal fluorescent probe should have well-separated absorption and emission spectra, a not too large emission spectrum (to avoid overlap), a quantum efficiency near unity (with the absence of nonradiative de-excitation processes), a large extinction coefficient (to avoid high excitation powers) and a perfect photostability. An in-depth description of the physical properties of fluorophores is beyond the scope of this article and can be found here [20-22].

For live cell imaging, the fluorescent probes should also penetrate in the cell without causing damage. Probe entry mechanisms include esterification of dyes to promote their cellular uptake, the use of synthetic vesicles containing probe [23], and mechanical techniques such as microinjection and electroporation.

Fluorescent proteins

The discovery of the naturally occurring green fluorescent protein (GFP), which was first isolated from jellyfish, revolutionized live cell imaging. It emits green fluorescence light under blue light excitation. It was discovered by Osamu Shimomura, in 1962, who purified it from the Aequorea victoria jellyfish [24]. It was only in 1992 that Douglas Prasher cloned the sequence of GFP and Martin Chalfie expressed this sequence in vivo [24]. Later on, the group of Roger Tsien reported the first crystal structure of GFP, showing the way for the creation of GFP mutants, to finally obtain different color variants and improve the fluorescence signal and photostability [25-27]. Shimomura, Chalfie, and Tsien were awarded in 2008 with the Nobel Prize in Chemistry for their work on the genetically encoded fluorescent proteins.

Mutation of the GFP gene led to the development of blue fluorescent protein (BFP) and a cyan fluorescent protein (CFP), which have altered absorption and emission properties from wild-type GFP [28]. Yellow fluorescent protein (YFP) and DsRed fluorescent protein derivatives have varied emission properties and thus allow scientists to use several fluorescent protein probes within the same cell. For example, a red fluorescent protein tdTomato transgenically expressed in neural stem and progenitor cells was used to delineate the neurogenesis in adult mouse brains [9].

The reason why GFP and fluorescent proteins (FPs) became so popular in biology is that with no external labeling, one can label cell compartments, or even whole cells, tissues or organisms, with the appropriate introduction of the FP gene in the genetic material of the cell or organism under study. For cell labeling, for example, it is sufficient to introduce in the cell nucleus a recombined gene including the FP encoding sequence combined with the sequence coding for the protein gene of interest. A mutant of the desired protein will then be expressed and will have fluorescence properties while keeping its functional properties, allowing highly specific labeling and thus analysis of the cellular structures and dynamics with imaging. Commercial expression vectors are available to label specific structures and organelles in live cells. For example, Choi JH et al used CellLight ER-RFP baculovirus from Life Technologies to label endoplasmic reticulum [29]. Vodnala SK et al monitored the autophagy flux of live T cells transduced with LC3-mcherry-eGFP constructs [30].

The FPs do not have to be expressed in tandom with the target protein. The target protein can be expressed with a peptide/protein tag, or even "spaghetti monster" tags [31], and be detected through the expression of, for example, anti-tag scFv, linked with an FP [32], or in the case of F-actin, a LifeAct peptide [33, 34].

Although convenient for intracellular bio-imaging, GFP and its derivatives lose their fluorescence properties under a continuous illumination period. Great attention should be paid to the expression levels of the FPs, that should be kept at similar levels than the ones of the endogenous proteins under study. Besides, when choosing a specific FP, one has to take into account the tendency of the FPs to aggregate and dimerize, which depends on various parameters, like temperature or cell line. The maturation time of the fluorophore should also be considered and varies between FPs. Zhao N et al discussed the three caveats of GFP: 1) long fluorophore maturation times, 2) slow GFP maturation times, 3) large, permanently attached, and dim moiety, which prevent its applications in particular areas of research, and designed an scFV against the HA tag as an alternative probe [32].

More recent developments include photoactivatable, photoconvertible or photo-switchable fluorescent proteins such as PA-GFP, mEOS or Dronpa [35]. PA-EGFP brightens 100 times with UV illumination, mEOS is switched from green to red with UV excitation and Dronpa is a reversibly switchable green FP that can repeatedly be turned ON with UV light (400nm) and rapidly photoswitched OFF with an excitation wavelength of 490 nm.

Organic fluorophores

The second family of fluorescent probes are organic fluorescent molecules. They can be coupled explicitly to biomolecules (peptides, proteins, oligonucleotides) or cell compartments in two different ways: i) by covalent coupling (direct chemical reaction), ii) by an intermediate attachment with a peptide or antibody, which is specific to the biomolecule. The most common probes are organic dye molecules, which can be synthesized and are commercially available. They are widely used in routine procedures and well-established protocols [36]. Comparing to fluorescent proteins, they have a wider spectral range, greater photostability, smaller size and most often are brighter.

A big family of organic dyes are the sulfonated rhodamine derivatives, termed Alexa Fluors, which have intense emission, greater photostability, and increased water solubility than the traditional first developed rhodamine derivatives and have been commercially provided by Molecular Probes (Alexa Fluor is a registered trademark of Molecular Probes) [37]. Alexa Fluor dyes range in excitation and emission wavelengths, as indicated by their alphanumeric names [38]. There are many variants so that multiple labeling experiments can be done exclusively with these dyes. Alexa Fluor dyes are available both as reactive intermediates (maleimides, succinimidyl esters, hydrazides), and pre-conjugated to a variety of other molecules, including phalloidin, dextrin, and many secondary antibodies.

Another prominent family of stable dyes are the Cyanine, including Cy2, Cy3, Cy5, Cy7 and their derivatives, which are commercially available in reactive dye form or coupled to other molecules [39].

Labeling of proteins with organic dyes in living cells can be possible either by direct or indirect labeling. Direct labeling targets most often amine and cysteine groups. Frequently, the labels are incorporated at the N-terminus or Cys/Lys side chains of the peptide [40, 41]. Indirect labeling consists of using peptide tags or protein tags conjugated to organic fluorophores.

Peptide tags

When using peptide tags, either the peptide tag is labeled and then attached to the protein through ligation, or it is genetically engineered onto the target protein and then are reacted with the corresponding fluorophore. This approach is useful for proteins sensitive to internal labeling or mutagenesis. As reviewed by O’Hare et al [42], some of the peptide solutions rely on enzyme-catalyzed fluorescent labeling (i.e., ACP tag [43] ), or simple chelation of fluorescent substrates (i.e., FlAsH labeling [44] ).

A typical case of peptide labeling is biotinylation by BirA, which covalently attaches biotin to lysine in a peptide tag. The peptide tag contains the biotin recognition sequence and can be ligated to the protein [45, 46]. BirA can also be expressed separately in processes like dimerization-induced quantitative BioID (DiQ-BioID) [47]. Biotin can also be purchased with reactive groups to target thiol or amine groups [48], but the specificity is lower compared to BirA.

Peptide fluorescent tags are small in size, which helps prevent the alteration of a target’s natural behavior.

Protein tags

The other labeling strategy is using protein tags, such as the self-labeling proteins (also called suicide enzymes) like SNAP-, CLIP-, or Halo-Tags, which can transfer a chemical group from a substrate to themselves [49-52]. Chromophores have been developed to detect protein aggregations between individual proteins and between two proteins through Halo-tag [53] and/or SNAP-tag in living cells [54, 55].

The possibility to add various fluorophores to their specific substrates opens a wide range of possible labeling. The self-labeling protein methods are very efficient to label membrane proteins fused to the self-labeling protein on their extracellular terminus and have been successfully used on G protein-coupled receptors [56, 57], tyrosine kinase receptors, ionic channels [58] or even intracellular proteins like histones [59].

Their main advantage is that they enable stable, covalent, and specific labeling of membrane receptors. However, potential background fluorescence and unspecific sticking of hydrophobic fluorogenic molecules to cell membranes may limit the usefulness of these systems for live cell imaging.

Live Cell Imaging figure 1
Figure 1. An example of fluorescently labeled mitochondria from Figure 1 in [1]. Fluorescent microscopy images are on the left, and bright field microscopy images are on the right. Live cell mitochondrial staining exhibits high mitochondrial reactivity in myotubes but not undifferentiated cells. C2C12 cells were seeded on the cell culture dishes. Upon reaching confluence, the cells were transferred to differentiation media (DM) to induce myotube (MT) formation. The DM was changed every two days. MitoTracker and Hoechst were added into the media for fluorescence staining of mitochondria and nuclei. After 30 min of incubation at 37C, live-cell images were recorded by bright field or fluorescence microscopy at day2 (A) and day4 (B). The cells were kept for 4days in DM with/without Hoechst 33342 (1 mM), and then Hoechst was added to stain nuclei (C). Cell morphology was also recorded by bright-field phase-contrast microscopy. MitoTracker and Hoechst signals are shown in red and blue, respectively (scale bar in A = 10 mm, B = 100 mm, and C = 50 mm). doi:10.1371/journal.pone.0028628.g001

MitoTracker and MitoFluor target mitochondria and are trapped in the organelles after chemical modification (oxidation) [60]. Figure 1 provides an example of MitoTracker staining of a muscle cell line [1]. Herb M et al stained mitochondria in cultured peritoneal macrophages with mitochondrial membrane potential-sensitive dye MitoTracker Red CMXRos from Thermo Fisher Scientific [61], so did L Schaupp et al for dentritic cells [62]. LysoTracker [63] and LysoSensor probes target lysosomes, and BODIPY derivatives are commonly used to mark the Golgi apparatus. A turn-on ESIPT probe has been identified for the detection of homocysteine and cysteine in water with live cell-imaging [64] and so has a fluorescent sensor for lead and aluminum [65] and for fluoride [66].

The domain of organelle-specific dyes is under constant development, with probes even brighter, smaller and more specific. For instance, the Nile-Red lipid droplet marker that is widely used until now could be soon replaced by the recently developed StatoMerocyanines (SMCy) probes, which present narrow absorption and emission spectra. They are based on an indolenine moiety and a dioxaborine barbiturate derivative. They fluoresce from yellow to the near-infrared and display remarkably high molar extinction coefficients (up to 390 000 M-1 cm-1) and high quantum yield values (near to 1). All the members of this new family specifically stain the lipid droplets in live cells and tissues with very low background noise as recently shown [67].

Indicator probes

Fluorescent probes designed to interact with inorganic ions, metals, thiols, and sulfides are also well utilized in live cell imaging. These probes, known as spectrally sensitive indicators, bind a target ion with specificity, which causes a change in their fluorescent properties. Many indicator probes, such as Ca2+ sensors, like Fluo-4 AM [68], fura-2, indo-1, and rhod-2 [69, 70] or Oregon Green 488 BAPTA-1 AM from Life Technologies [71], Cal-520-AM from AAT Bioquest [72], have been developed to interact with calcium and can measure both localized calcium concentrations within cell compartments, and quantitative changes during calcium flux in the cytoplasm [73].

Magnesium, sodium, potassium, and zinc are some other ions are that can be measured with specific fluorescent indicators. Cyclic AMP can be measured through cAMPr [74].

Inorganic fluorescent probes

During the past 20 years new inorganic fluorescent probes have been proposed, as alternatives to organic fluorophores, with the aim to achieve higher fluorescence yield and lower photobleaching. Inorganic fluorescent nanocrystals include semiconductor nanocrystals (also called Quantum Dots, QDs) [75], silicon nanoparticles [74], lanthanide-doped oxide nanoparticles [76] and fluorescent nanodiamonds [77, 78]. Some of the above systems are already commercialized and widely used, some others are new and under development.

Live Cell Imaging figure 2
Figure 2. A diagram of a quantum dot, and images utilizing quantum dots from figure 4 in [2]. Targeting of ES cells by peptide-conjugated quantum dots. (A) Scheme of the peptide-conjugated quantum dots. (B) Bright-field and fluorescence microscopy of APWHLSSQYSRT peptide-conjugated quantum dots targeting ES cells. Dashed circle shows an ES cell colony. The fluorescence signal of peptide-conjugated quantum dots is shown in red and cell nuclei are shown in blue (Hoechst 33258). PMEF cells and quantum dots without conjugation of peptides were set as the controls. Scale bar, 100 mm. (C) Fluorescence microscopy of APWHLSSQYSRT peptide-conjugated quantum dots with mouse ES (mES) cells. The binding of APWHLSSQYSRT peptide-conjugated quantum dots to mES cells were undetectable. Free quantum dots without conjugation of peptides were set as the control. Scale bar, 100 mm. doi:10.1371/journal.pone.0012075.g004

The most widely used QDs are CdSe and CdTe particles (II/VI family), which are commercially available (Invitrogen, Evident Technologies, etc.). The core size of typical QDs is 2-10 nm. A shell, usually made of ZnS is necessary to stabilize the fluorescence properties. For their solubilization, the particles are further coated with various polymers, which have the necessary chemical groups for biomolecules attachment. In the end, the total size of the particles is in the range of 20-50 nm. This is one or two orders of magnitude larger than the equivalent system using a single dye molecule as the fluorophore but is still in the nanometric scale. Their fluorescence signal is brighter than the equivalent of organic dye molecules, and they present a better photostability allowing observations and tracking over an extended period. They often coupled to antibodies, proteins, or carbohydrates for fluorescent imaging applications [79, 80]. Figure 2 provides an example of quantum dot imaging [2].

QDs may appear cytotoxic on long term due to their core chemical composition, composed of cadmium, a very toxic heavy metal element [81, 82]. Little is known yet on this issue, and further investigations are necessary.

TypeCommon ApplicationProCon
Fluorescent Proteins (FP)Targeting probe to specific cellular compartments or to interact with particular proteins
FRET
Direct in situ labeling
Can be attached to a target genetically
Very specific
Low toxicity
Large Size
Beta barrel shape may hinder molecules under study
Photobleach
Low signal intensity
Fluorescent PeptidesSimilar applications as FP but utilized when size of FP may hinder studySmall size
Low Toxicity
Targets Specific Interactions
Often requires coupling to molecules
Organic FluorophoresCan target cell structures
Label mRNA sequences
Small
Good for multiplexing
Limited photostability
Wide range of colors
Require coupling to another molecule
Photobleach
Hydrophobic
Somewhat toxic
Lower specificity
Indicator ProbesIndicate presence of ions and metalsVery specificOften requires coupling to molecules
Quantum DotsLong term imaging studies
Real-time tracking over extended periods
Very photostable
Good for multiplexing
Intense signal
Enable very sensitive detection
Coupling to another molecule necessary
Interference
Table 3. Live imaging probes and their applications.
Bioluminescence tags

The drawbacks of the standard fluorescent reporters include photosensitivity and autofluorescent background. Bioluminescence, which does not require an additional light stimulation, may also be effectively applied for live-cell imaging. Bioluminescence imaging may be performed by the expression of luciferases in various cell types. Noninvasive analysis of the cells expressing luciferase is often used in preclinical studies [83]. Moreover, multicellular imaging may be done using structurally different luciferins and luciferases. For example, firefly, Renilla, and Vargula hilgendorfii luciferases were used to analyze cellular responses in a mouse tumor model [84]. Besides, luciferins may be modified to enhance luminescence detection. In particular, modified luciferins, such as CycLuc1, were shown to have increased membrane permeability [85]. Also, a recently engineered subtype of the firefly enzyme was demonstrated to interact with luciferin substrate more effectively than its older analogs [86].

Another luminescent reporter, Nano-lantern, was used for imaging of cellular organelles in vivo [87]. Several color subtypes of NanoLuc-based Nano-lantern have recently been developed [88]. Besides, another new luminescent protein Antares, which is significantly brighter than Nano-lantern, produces strong photon emission and can be used in different tissues [89] and so does its derivative Antares2 [90].

Others

Finally, a variety of cell-permeable fluorescent molecules are available that more or less specifically label intracellular organelles. The fluorophore is coupled to a targeting moiety that will localize it to the target organelle. For nuclear staining, most common probes for living cells are intercalating DNA dyes such as DAPI, Hoechst (blue) and Draq5 (far-red).

Recently, a carboxylated silicon–rhodamine (SiR) derivative of Hoechst dye (named SiR–Hoechst or SiR-DNA from Cytoskeleton Inc [13] ) has been synthesized, and found to have a much lower KD for DNA binding (8.4μM), about 1,000-fold lower than that of Hoechst 33342 for DNA, and it has no detectable affinity towards double-stranded RNA. The dye is excited at 640 nm, and its fluorescence intensity at 670nm increases about 50-fold upon DNA binding. SiR derivatives exist in equilibrium between nonfluorescent spironolactone OFF state and a fluorescent zwitterion ON state. Aggregation of SiR or unspecific binding to hydrophobic surfaces favors the OFF state. Their interaction with polar protein surfaces switches the fluorophores into the ON state. The SiR conjugated to ligands that bind to the nucleus (SiR-Hoechst) or other organelles, like actin or microtubules. Stimulated emission depletion (STED) microscopy is shown for this kind of probes [91, 92], of which the photodamage on live cells have been evaluated [93]. Wang H et al observed the protection of target-bound dCas9-gRNA complex in the cellular environment and designed CRISPR LiveFISH procedure to label DNA and RNA in cells in real time [94]. Hafner AS used STED microscopy to observe vGLUT1+ sorted synaptosomes to study the local protein synthesis in neuronal pre- and postsynaptic compartments [95].

On a side note, probes such as To-Pro-3 [70, 96] can be used to assess/counterstain dead or fixed cells.

Microscopy

Light microscopy is at the heart of live cell imaging. Light microscopy requires light sources, a microscope objective (most often on a microscope stand) and a detector (camera or point detector). Many different systems have been employed. Gaglia G et al, for example, took time-lapse images of cultured cells under treatments with GE IN Cell Analyzer 6000, Perkin-Elmer Operetta High-Content Imaging System or Nikon Eclipse Ti Live-Cell Imaging System [15]. Multi-mode readers such as Cytation 5 from BioTek have been used [97].

Transmission light microscopy

The first applied light microscopy method was the wide field transmission microscopy. The light is passed through a condenser to focus it on the specimen to get a maximum illumination. After the light passes through the specimen, it goes through the objective lens to magnify the image of the sample and then to the occulars, where the enlarged image is viewed or to the detector. Transmission light microscopy falls into four families i) bright field, ii) dark field, iii) phase contrast (PH) and iv) differential interference contrast (DIC) microscopy.

Bright field microscopy

Here the sample is not stained, and the transmitted light is detected, for example, as with Leica SP8 bright-field microscopy [98]. Setting up the light path is essential. The most used method is the Koehler illumination (August Koehler was the inventor). It employs both a field and aperture iris diaphragm to set up the illumination beam. The condenser is used to focus parallel rays on the specimen, giving the advantages of an evenly illuminated field with minimum heating of the sample. Staining can be used to enhance contrast (most common is toluidine blue, for a semi-thin section of all types of tissues).

Dark field microscopy

The specimen is illuminated from the side. The scattered light from the sample is detected through the objective and is seen against a dark background, with improved contrast. A special darkfield condenser is needed to provide the oblique illumination.

Phase contrast microscopy

Here the specimen is illuminated by a hollow cone of light passing through a phase annulus in the condenser. Special phase contrast objectives are used that have integrated a corresponding phase plate. Light passing through the sample is slightly retarded compared to rays that do not pass through, and the phase plate achieves further retardation. Destructive and constructive interferences occur of rays that combine after the objective, which produce bright and dark feature in the image. It is a widely used technique, especially for big structures (nuclei, organelles) and is under continuous development [99].

Differential interference contrast (DIC) microscopy

DIC microscopy requires a polarized light source and two polarizing filters, and two special Wollaston prisms that split the light into two beams. Illumination polarized light is first separated by the first prism into two beams, then passes through the sample (one beam can pass through the sample and the other next to it, thus creating a path and so a phase difference between the two beams) and is recombined by the second prism after the objective before passing through the second polarization filter and been detected. The result is a 3D-like image with shades of grey, giving the impression of al relief in the image [100].

Live Cell Imaging figure 3
Figure 3. A 3D fluorescent confocal microscopy image and the 3D image generated as presented in figure 1 in [3]. Trypsinised SW480APC cells stained with Calcein AM fluorescent dye (green), Hoechst 33342 nucleic acid stain (blue) and Vybrant DiI cell-labeling solution (red), fluorescent marking the cytoplasm, nuclei and membrane respectively. Different overlayed fluorescent signal views of a sample of the acquired image stack are as shown, namely (A) 2D sectional view; (B) orthogonal views of selected cells; (C) 3D volumetric view with blending effect. doi:10.1371/journal.pone.0031882.g001
Fluorescence microscopy

As described previously in the review, fluorescence can be used as a label when preparing biological samples. The fluorescent molecules (fluorochromes) after excitation emit light at a different (longer) wavelength. Fluorescence microscopy employs reflected rather than transmitted light. Transmitted light techniques can very well be combined with fluorescence microscopy. Filters are necessary to visualize the fluorescence emitted. Most often a dichroic mirror cube is used that comprises the dichroic beam splitter and an excitation and emission filter. The dichroic mirror reflects shorter wavelengths and lets passes longer wavelengths of light to isolate the excitation and emission wavelengths.

A bright source is also necessary for the fluorochrome excitation. For wide field microscopy, it can be a mercury lamp, or LED sources. For confocal microscopy, monochromatic laser sources are used.

An advantage of fluorescent microscopy over bright field, dark field, and phase contrast microscopy is that different fluorescent dyes can be used simultaneously to stain different components of the cell, and can be detected simultaneously.

Widefield microscopy

This is the most well-known technique of fluorescence microscopy, where the microscope forms an image of the sample on the detector (camera).

Live cells are most commonly imaged on an inverted epifluorescence microscope. In such a microscope, the objective images the sample from below. Inverted microscopes allow imaging through a glass coverslip. Cells can be grown in coverslip-bottom Petri dishes or multiwell plates containing growth media.

Confocal microscopy techniques

A significant limitation of conventional wide-field microscopy is that not only is the plane of focus illuminated, but much of the specimen above and below this point is also illuminated resulting in out-of-focus blur. This leads to a reduction of contrast and resolution, especially for thick samples, where the effect is more dominant.

For samples that are not too thick and not too densely labeled with fluorophores, this out of focus light is not a significant problem. However, for thick, densely stained samples or in cases in which we wish to achieve well-resolved 3D images, this out-of-focus light can obscure valuable information.

Confocal microscopy suppresses the out-of-focus structures at image formation. The sample is illuminated by a focused laser beam at a single point in the focal plane, instead of a full wide-field illumination [101]. The sample is raster-scanned by using galvanometric mirrors. Light from the excited point is detected on a point detector (most often photomultiplier – PMT) after passing through a pinhole. The pinhole does not permit rays of light from out-of-focus planes to pass. The wavelength of light, the numerical aperture of the objective and the diameter of the diaphragm (wider detection pinhole reduces the confocal effect but increases the detected signal) affect the depth of the focal plane.

Confocal microscopes, for example, LSM 710 META laser scanning confocal microscope [18], perform optical sectioning, a process that allows the system to acquire in-focus images from several depths within a sample and then reconstruct images taken in a point-by-point manner that can be merged to create a 3D representation of a sample. Figure 3 presents an example of three color fluorescent confocal microscopy, and the 3D image generated [3].

Spinning disk multifocal confocal microscopy was developed to overcome the time-consuming point-by-point raster scanning of the confocal microscopy [102], for example, Olympus IX81 inverted spinning disc confocal microscope [14], or CV1000 automated spinning-disk microscope [103], or a Yokogawa spinning-disk confocal on a Nikon Ti inverted microscope [104]. It uses a disk of pinholes and a disk of micro-lenses from which the laser passes through. The pinhole and lens disk turn very fast (1000-10000rpm) which significantly increases the acquisition speed. The detector here is a camera, and the technique is widely used for live cell imaging. Confocal imaging provides several advantages over traditional wide-field. It shows an increased contrast and resolution and enables imaging of thicker sections, limiting the out-of-focus signal.

Specific advanced microscopy approaches

Advanced microscopy approaches have been developed for live cell images. They are discussed below.

Multiphoton microscopy

Multiphoton microscopy is a type of laser-scanning microscopy that involves nonlinear excitation that elicits fluorescence in a thin plane, which improves the 3D imaging, reduces excitation of out-of-focus planes and acquires more fluorescence signal (no need of a pinhole, more scattered photons are detected) [105].

Multiphoton microscopy can image hundreds of microns deep within a section and is valuable for the study of thick sections, tissues, and even animals. Second harmonic generation (SHG) microscopy can be combined with this technique. In SHG microscopy two photons are upconverted to exactly twice the frequency of the excitation laser. SHG creation requires a noncentrosymmetric assembly, limiting the use of SHG to imaging collagen, myosin and microtubule assemblies. The main advantage though is that no labeling is needed, facilitating the imaging of these structures in live cells, tissues, and animals.

TIRF microscopy

Total internal reflection fluorescence microscopy (TIRFM) selectively excites fluorophores in a specific region of a specimen that is adjacent to the glass-water interface [106]. It relies on total internal reflection of a laser beam at the interface of the coverslip and the aqueous sample. The reflected laser beam sets up an evanescent wave that penetrates in the sample only a few hundreds of nanometers (the penetration depth depends on the incident angle of the laser beam). Only fluorophores in that depth are excited and thus emit light. No background is then detected of the rest of the sample. The low background level facilitates the observation in time of single molecules [107].

This technique is well used in studies of the plasma membrane, such as studies examining the process of exocytosis or the diffusion of membrane receptors [108, 109] or chromatin dynamics [53].

Live Cell Imaging figure 4
Figure 4. Visualization of the Ca2+-dependent interaction of hEAG1-Cerulean with CaM in cells by FRET from figure 1 in [4]. Fluorescence intensity and FRET efficiency (E%) images of HEK-293 transfected with (A) the C-terminus of hEAG1 or (B) the full-length hEAG1 labeled with Cerulean (donor) and calmodulin (CaM) or apoCaM labeled with YFP (acceptor). Cells were stimulated with 1 mM ionomycin in the presence of 1.8 mM Ca2+ (+Ca2+) or 1 mM EGTA (-Ca2+) for 10 min. FRET was measured by acceptor photobleaching. Bleached areas are indicated by white boxes. FRET efficiencies are shown in pseudo-color. Scale bars: 5 mm. doi:10.1371/journal.pone.0010873.g001
FRET microscopy

Forster resonance energy transfer (FRET) microscopy was first formalized by Theodor Foerster in the middle of the twentieth century [110]. FRET strategies are based on a non-radiative resonance energy transfer between a donor and an acceptor. The efficiency of the transfer depends on the energy compatibility between the donor and the acceptor (importance of spectra overlap), the orientation of the probes (the maximum energy transfer occurs when the transition dipole moments are parallel) and the distance between the probes (energy transfer occurs only if the fluorophores are in a proximity, lower than 10nm, and the efficiency is inversely proportional to the sixth power of the distance). The technique is considered as a strong indicator of strong interactions of molecules, as energy transfer of the donor to the acceptor occurs only if the probes are in a very close distance.

Enhanced CFP and YFP proteins are often used as a FRET pair in imaging studies. The FRET phenomenon can be utilized for biosensing by engineering a donor and acceptor into a fluorescent protein (FP) that interacts with a target molecule whose conformational change is under study. When the target molecule is stimulated and its conformation changes, the donor and acceptor get close enough to cause the FRET signal. Biosensors using this approach have been developed for a wide array of molecules, including calcium [111], proteases [112], cAMP [113], and kinases [114]. See Figure 4 for an example of FRET images from [4].

The FRET analysis is either made by the intensity ratio of donor and acceptor or by calculating the change of the probes lifetimes due to the transfer by FLIM technique. FRET presents some drawbacks the direct excitation of the acceptor at the donor excitation wavelength, the emission of the donor at the emission wavelength of the acceptor, the dynamic FRET resulting from the random collision of donor and acceptor diffusing in the medium. Special care should thus be taken when planning (choice for a donor-acceptor pair), performing and analyzing FRET experiments.

Live Cell Imaging figure 5
Figure 5. An example of FRAP images and charts plotting intensity versus time from figure 2 in [5]. A-E: snapshots of a cell during data acquisition; A: the cell before photobleaching (panel labeled “pre”); B-E: the cell during recovery after photobleaching (timestamp indicates the time after the photobleaching pulse). Dotted circle indicates the bleaching spot (B). Scale bar 2 µm. F–J: the fluorescence intensity along the dotted cross-section at given time points (corresponding to images on the left: A–E), where black is the normalized fluorescence intensity and red is the fit. K, L: the change of intensity in the course of the recovery at (K) the bleached pole of the cell (R1) and at (L) the opposite pole of the cell (R2). Black squares show the normalized data points and red lines the corresponding fits. doi:10.1371/journal.pone.0025664.g002
FLIM microscopy

Fluorescence Lifetime Imaging Microscopy (FLIM) allows visualization of the lifetime of the excitation state of a fluorescent molecule [115]. FLIM can distinguish a FRETing donor from a non-interacting donor based on lifetime excitation and can increase the signal/noise ratio of FRET [116]. With FLIM microscopy one can also measure with high precision the changes of the lifetime of the labeled proteins, changes that are directly linked to their environment.

Techniques Advantages Drawbacks Perspectives
Transmission Light MicroscopyBright-FieldEasy to useLow contrast Use of new equipment (detectors) and update setups to increase contrast and image smaller structures
Dark-FieldImproved contrastNeeds special condenser
Phase contrastHigh contrastNot efficient for all structures
DICRelief imageNeeds specific alignment – prone to artifacts
Classic wide-field fluorescence microscopyEpifluorescence microscopyEasy to use
Allows fast imaging
Photobleaching
Detects out-of-focus signal
Combine it with other microscopy techniques
Confocal microscopyLaser-scann confocal3D sectioning
Thicker samples
Time-consumingFaster techniques are implemented like fast-Airy scan
Spinning disk3D sectioning
Thicker samples
Fast imaging
PhotobleachingImprove illumination homogeneity
Wider spinning disk heads and wider camera chip
Advanced techniquesMultiphoton Microscopy3D sectioning
In-depth imaging
Time-consuming
Destroy sample if high power used
Faster scanning with resonant scanners or AOD solutions
Use of lasers with longer wavelengths to broaden the excitation range
TIRFOptical sectioning for structures near to coverslide-water interface (<200nm)Restricted to coverslide-water interface imaging
Difficult to quantify excitation
Super-critical angle fluorescence for more confined excitation
FRETDirect label with fluorescent proteins
Detect direct interactions (<10nm)
Analysis artifacts linked to donor-acceptor caracteristicsDevelop new donor-acceptor pairs
Perform FRET with advanced microscopy methods to avoid artifacts (FLIM or spectral detection)
FLIMMeasure with high precision lifetimes or proteins and influence of environmentTime-consumingImplement new detectors for faster FLIM imaging
BRETHigher signal specificityNeeds long acquisition time per frame to collect signal – risk of detecting increased noise levelsDevelopment of brighter luciferases
FRAPImage protein diffusion in cellsMeasurement averaged over numerous fluorescent proteinsImplementation of new detectors for more sensitive monitoring of dynamics
FCSAdditional information for protein dynamics in shorter timesUnable to characterize direct interactions due to diffraction limitCombine FCS with super-resolution techniques to decrease the excitation volume
SPTTracking of individual protein diffusion
Improved localisation precision
Adapt fluorescent labeling/densityUse brighter probes to increase the signal to background ratio and thus the precision
Combine SPT with PALM
PALMStudy protein organisation and diffusion in dense samplesPhotophysical artifacts (blinking) If not used in SPT mode, technique is slow for live imagingUse new brighter and more stable probes
Light Sheet MicroscopyIsotropic 3D resolution
Low background
Adapted for thick sample live imaging
Illumination artifacts
artifacts for obtaining the 3D final image through fusion
New excitation modes proposed
New algorithms and methods for more efficient fusion
Table 4. Overview of live cell imaging techniques.
BRET microscopy

BRET similarly to FRET relies on a transfer of energy, the donor probe being replaced by a complex formed by a bioluminescent enzyme, usually Renilla luciferase (Rluc), and its substrate coelenterazine (clz). Donor excitation is achieved with a chemical substrate instead of light excitation, therefore preventing any undue excitation of the acceptor.

The first BRET donor was the complex Rluc/clz (emission peak at 480 nm), and YFP was the acceptor (emission peak at 525 nm) [117]. Because of the significant overlap of Rluc and YFP emission spectra, BRET intensity is determined by calculating the BRET ratio, a mathematical calculation allowing the subtraction of donor emission at the acceptor wavelength [118]. Optimization of the enzyme/substrate complexes and use of new acceptors to reduce emission spectral overlap have been carried out, and variants of BRET have been implemented [119, 120]. Recently the smaller luciferase (Nluc) and the Antares2, described being much brighter than Rluc8 has proven to significantly enhance the resolution of BRET imaging [90, 121].

FRAP approach

Live cell imaging is often used to observe the movement of molecules within cells. Several microscopy techniques have been developed to do this. Fluorescence recovery after photobleaching (FRAP) is one such procedure [17, 18]. With FRAP, a subcellular region is photobleached by exposing it to high intensities of light and the fluorescence recovery is monitored over time. This happens at different rates and with varied completion depending on the amount of diffusion or active transport of the fluorescent molecules into the bleached area [122]. Zhao N et al used FRAP to evaluate the binding kinetics of HA epitopes with a cognate scFv inside living cells [32]. An example of conventional FRAP is presented in Figure 5 [5]. Another application of FRAP is to study the movements of molecules through gap junctions [123]. Fluorescence loss in photobleaching (FLIP), is similar, but an area is continuously photobleached [124].

FCS spectroscopy

Fluorescence correlation spectroscopy (FSC) is used to gather information about the diffusion rate and concentration of target molecules by measuring the fluorescence fluctuation in a small volume for short periods [125].

SPT approaches

Single-molecule tracking (SPT) identifies fluorescence spots corresponding to single fluorophores, pinpoints their localization with high precision and follows their diffusion in time [126]. It is lately widely used to study dynamics of proteins, like motor proteins like kinesin and dynein [127] the spatiotemporal dynamics and cytoplasmic assembly of the 26S proteasome [128] or dynamics of GPCR receptors [109].

However, if SPT allows an increase in spatial resolution, as the single fluorophore can be pinpointed to the center of mass of its diffraction-limited spot, the resolution achieved in these experiments is not higher than 20–30 nm, a value depending on fluorescence intensity, receptor density, and temporal resolution.

PALM microscopy

New techniques have recently been adapted to overcome receptor density, such as photo-activation light microscopy (PALM), using photoactivatable fluorophores. PALM is based on the photoactivation or photoswitching and subsequent bleaching of sparse fluorescent proteins in the sample, thus separating the molecules temporally and distinguishing them in space (localization procedure similar to the one for SPT) [129]. sptPALM is similar to the SPT method without the limit of protein density. It is often combined with TIRF, to increase the signal to background ratio and thus increase the localization precision. PALM is a handy tool for studying protein organization in dense samples. However, potential photophysical artifacts can influence the results. For example, blinking molecules, which can be counted more than one time and consequently be interpreted as a cluster, are a common issue for this technique. Performing control experiments is of crucial importance for quantitative PALM measurements.

Light sheet microscopy

For live imaging of thicker samples, new microscopy approaches exist, such as Light Sheet Microscopy. The sample is illuminated from a plane orthogonal to the imaging plane, eliminating the out-of-focus light as only light from the focal plane is excited. Thanks to the selective illumination of the plane, the total light exposure is reduced, and so are the photobleaching and phototoxicity. The sample can rotate to perform whole sample illumination, and a fusion process is carried out at the end of the acquisition for a 3D image of the sample with an isotropic resolution [130]. Double illumination schemas exist to increase speed and avoid illumination artifacts. The technique has been recently coupled to super-resolution microscopy to offer fast and super-resolved imaging of living cells [131]. Serra D et al custom-developed a light sheet microscopy to image the development of intestinal organoids [132].

Analysis

Image analysis is critical to each microscopy technique described above and is well reviewed here [133]. The primary goal of image processing is to convert the image into useful information. This focuses on three aspects: reducing background noise, enhancing contrast, and quantifying the intensity of a signal. Many software programs designed to analyze and annotate the image data are available, and many of them are open source [134, 135], for example, ImageJ and Icy [18]. Advanced algorithms for image processing and data analysis are available and are quite specific to the type of image and sample being studied. While optimizing image quality and resolution during image analysis, it is important to remember that there are ethical guidelines for manipulating digital images responsibly, so that they continue to reflect the raw information embedded within them [136].

Declarations
Revision history

Jennifer Walker-Daniels wrote the article in 2012, and Orestis Faklaris made significant revision in 2018.

References
  1. Miyake T, McDermott J, Gramolini A. A method for the direct identification of differentiating muscle cells by a fluorescent mitochondrial dye. PLoS ONE. 2011;6:e28628 pubmed publisher
  2. Lu S, Xu X, Zhao W, Wu W, Yuan H, Shen H, et al. Targeting of embryonic stem cells by peptide-conjugated quantum dots. PLoS ONE. 2010;5:e12075 pubmed publisher
  3. Tan C, Gardiner B, Hirokawa Y, Layton M, Smith D, Burgess A. Wnt signalling pathway parameters for mammalian cells. PLoS ONE. 2012;7:e31882 pubmed publisher
  4. Goncalves J, Stuhmer W. Calmodulin interaction with hEAG1 visualized by FRET microscopy. PLoS ONE. 2010;5:e10873 pubmed publisher
  5. Mika J, Krasnikov V, van den Bogaart G, de Haan F, Poolman B. Evaluation of pulsed-FRAP and conventional-FRAP for determination of protein mobility in prokaryotic cells. PLoS ONE. 2011;6:e25664 pubmed publisher
  6. Jost A, Waters J. Designing a rigorous microscopy experiment: Validating methods and avoiding bias. J Cell Biol. 2019;: pubmed publisher
  7. Freeman S, Uderhardt S, Saric A, Collins R, Buckley C, Mylvaganam S, et al. Lipid-gated monovalent ion fluxes regulate endocytic traffic and support immune surveillance. Science. 2020;367:301-305 pubmed publisher
  8. Gabriele M, Brandão H, Grosse Holz S, Jha A, Dailey G, Cattoglio C, et al. Dynamics of CTCF- and cohesin-mediated chromatin looping revealed by live-cell imaging. Science. 2022;376:496-501 pubmed publisher
  9. Pilz G, Bottes S, Betizeau M, Jörg D, Carta S, Simons B, et al. Live imaging of neurogenesis in the adult mouse hippocampus. Science. 2018;359:658-662 pubmed publisher
  10. Thal D, Schultz C, Botez G, Del Tredici K, Mrak R, Griffin W, et al. The impact of argyrophilic grain disease on the development of dementia and its relationship to concurrent Alzheimer's disease-related pathology. Neuropathol Appl Neurobiol. 2005;31:270-9 pubmed
  11. Lowndes R, Hallett M. A versatile light microscope heating stage for biological temperatures. J Microsc. 1986;142:371-4 pubmed
  12. Klein I, Boija A, Afeyan L, Hawken S, Fan M, Dall Agnese A, et al. Partitioning of cancer therapeutics in nuclear condensates. Science. 2020;368:1386-1392 pubmed publisher
  13. Lin Z, Gasic I, Chandrasekaran V, Peters N, Shao S, Mitchison T, et al. TTC5 mediates autoregulation of tubulin via mRNA degradation. Science. 2020;367:100-104 pubmed publisher
  14. Zhang H, Emerson D, Gilgenast T, Titus K, Lan Y, Huang P, et al. Chromatin structure dynamics during the mitosis-to-G1 phase transition. Nature. 2019;576:158-162 pubmed publisher
  15. Gaglia G, Rashid R, Yapp C, Joshi G, Li C, Lindquist S, et al. HSF1 phase transition mediates stress adaptation and cell fate decisions. Nat Cell Biol. 2020;22:151-158 pubmed publisher
  16. Greenwood D, Dos Santos M, Huang S, Russell M, Collinson L, Macrae J, et al. Subcellular antibiotic visualization reveals a dynamic drug reservoir in infected macrophages. Science. 2019;364:1279-1282 pubmed publisher
  17. Yu H, Lu S, Gasior K, Singh D, Vazquez Sanchez S, Tapia O, et al. HSP70 chaperones RNA-free TDP-43 into anisotropic intranuclear liquid spherical shells. Science. 2021;371: pubmed publisher
  18. Garcia Jove Navarro M, Kashida S, Chouaib R, Souquere S, Pierron G, Weil D, et al. RNA is a critical element for the sizing and the composition of phase-separated RNA-protein condensates. Nat Commun. 2019;10:3230 pubmed publisher
  19. Frigault M, Lacoste J, Swift J, Brown C. Live-cell microscopy - tips and tools. J Cell Sci. 2009;122:753-67 pubmed publisher
  20. Fernandez Suarez M, Ting A. Fluorescent probes for super-resolution imaging in living cells. Nat Rev Mol Cell Biol. 2008;9:929-43 pubmed publisher
  21. Terai T, Nagano T. Fluorescent probes for bioimaging applications. Curr Opin Chem Biol. 2008;12:515-21 pubmed publisher
  22. Hanson G, Hanson B. Fluorescent probes for cellular assays. Comb Chem High Throughput Screen. 2008;11:505-13 pubmed
  23. Massignani M, Canton I, Sun T, Hearnden V, MacNeil S, Blanazs A, et al. Enhanced fluorescence imaging of live cells by effective cytosolic delivery of probes. PLoS ONE. 2010;5:e10459 pubmed publisher
  24. Prasher D, Eckenrode V, Ward W, Prendergast F, Cormier M. Primary structure of the Aequorea victoria green-fluorescent protein. Gene. 1992;111:229-33 pubmed
  25. Ormo M, Cubitt A, Kallio K, Gross L, Tsien R, Remington S. Crystal structure of the Aequorea victoria green fluorescent protein. Science. 1996;273:1392-5 pubmed
  26. Zhang J, Campbell R, Ting A, Tsien R. Creating new fluorescent probes for cell biology. Nat Rev Mol Cell Biol. 2002;3:906-18 pubmed
  27. Shaner N, Steinbach P, Tsien R. A guide to choosing fluorescent proteins. Nat Methods. 2005;2:905-9 pubmed
  28. Heim R, Prasher D, Tsien R. Wavelength mutations and posttranslational autoxidation of green fluorescent protein. Proc Natl Acad Sci U S A. 1994;91:12501-4 pubmed
  29. Choi J, Zhong X, McAlpine W, Liao T, Zhang D, Fang B, et al. LMBR1L regulates lymphopoiesis through Wnt/β-catenin signaling. Science. 2019;364: pubmed publisher
  30. Vodnala S, Eil R, Kishton R, Sukumar M, Yamamoto T, Ha N, et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science. 2019;363: pubmed publisher
  31. Viswanathan S, Williams M, Bloss E, Stasevich T, Speer C, Nern A, et al. High-performance probes for light and electron microscopy. Nat Methods. 2015;12:568-76 pubmed publisher
  32. Zhao N, Kamijo K, Fox P, Oda H, Morisaki T, Sato Y, et al. A genetically encoded probe for imaging nascent and mature HA-tagged proteins in vivo. Nat Commun. 2019;10:2947 pubmed publisher
  33. Riedl J, Crevenna A, Kessenbrock K, Yu J, Neukirchen D, Bista M, et al. Lifeact: a versatile marker to visualize F-actin. Nat Methods. 2008;5:605-7 pubmed publisher
  34. Krndija D, el Marjou F, Guirao B, Richon S, Leroy O, Bellaiche Y, et al. Active cell migration is critical for steady-state epithelial turnover in the gut. Science. 2019;365:705-710 pubmed publisher
  35. Lippincott Schwartz J, Patterson G. Fluorescent proteins for photoactivation experiments. Methods Cell Biol. 2008;85:45-61 pubmed
  36. W.T. Mason. Fluorescent and luminescent probes for biological activity. Academic Press, London, 2nd ed edition, 1999.
  37. Panchuk Voloshina N, Bishop Stewart J, Bhalgat M, Millard P, Mao F, Leung W, et al. Alexa dyes, a series of new fluorescent dyes that yield exceptionally bright, photostable conjugates. J Histochem Cytochem. 1999;47:1179-88 pubmed
  38. HAINAUX H. [A REFORM OF A REFORM]. Concours Med. 1963;85:4907-11 pubmed
  39. Ballou B, Fisher G, Waggoner A, Farkas D, Reiland J, Jaffe R, et al. Tumor labeling in vivo using cyanine-conjugated monoclonal antibodies. Cancer Immunol Immunother. 1995;41:257-63 pubmed
  40. Amitani I, Liu B, Dombrowski C, Baskin R, Kowalczykowski S. Watching individual proteins acting on single molecules of DNA. Methods Enzymol. 2010;472:261-91 pubmed publisher
  41. Galletto R, Amitani I, Baskin R, Kowalczykowski S. Direct observation of individual RecA filaments assembling on single DNA molecules. Nature. 2006;443:875-8 pubmed
  42. O Hare H, Johnsson K, Gautier A. Chemical probes shed light on protein function. Curr Opin Struct Biol. 2007;17:488-94 pubmed
  43. George N, Pick H, Vogel H, Johnsson N, Johnsson K. Specific labeling of cell surface proteins with chemically diverse compounds. J Am Chem Soc. 2004;126:8896-7 pubmed
  44. Griffin B, Adams S, Tsien R. Specific covalent labeling of recombinant protein molecules inside live cells. Science. 1998;281:269-72 pubmed
  45. Chen I, Howarth M, Lin W, Ting A. Site-specific labeling of cell surface proteins with biophysical probes using biotin ligase. Nat Methods. 2005;2:99-104 pubmed
  46. Howarth M, Ting A. Imaging proteins in live mammalian cells with biotin ligase and monovalent streptavidin. Nat Protoc. 2008;3:534-45 pubmed publisher
  47. Birnbaum J, Scharf S, Schmidt S, Jonscher E, Hoeijmakers W, Flemming S, et al. A Kelch13-defined endocytosis pathway mediates artemisinin resistance in malaria parasites. Science. 2020;367:51-59 pubmed publisher
  48. Elia G. Protein biotinylation. Curr Protoc Protein Sci. 2010;Chapter 3:Unit 3.6 pubmed publisher
  49. Juillerat A, Gronemeyer T, Keppler A, Gendreizig S, Pick H, Vogel H, et al. Directed evolution of O6-alkylguanine-DNA alkyltransferase for efficient labeling of fusion proteins with small molecules in vivo. Chem Biol. 2003;10:313-7 pubmed
  50. Juillerat A, Heinis C, Sielaff I, Barnikow J, Jaccard H, Kunz B, et al. Engineering substrate specificity of O6-alkylguanine-DNA alkyltransferase for specific protein labeling in living cells. Chembiochem. 2005;6:1263-9 pubmed
  51. Keppler A, Gendreizig S, Gronemeyer T, Pick H, Vogel H, Johnsson K. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat Biotechnol. 2003;21:86-9 pubmed
  52. Gronemeyer T, Chidley C, Juillerat A, Heinis C, Johnsson K. Directed evolution of O6-alkylguanine-DNA alkyltransferase for applications in protein labeling. Protein Eng Des Sel. 2006;19:309-16 pubmed
  53. Davidson I, Bauer B, Goetz D, Tang W, Wutz G, Peters J. DNA loop extrusion by human cohesin. Science. 2019;366:1338-1345 pubmed publisher
  54. Jung K, Kim S, Liu Y, Zhang X. A fluorogenic AggTag method based on Halo- and SNAP-tag to simultaneously detect aggregation of two proteins in live cells. Chembiochem. 2019;: pubmed publisher
  55. Liu Y, Wolstenholme C, Carter G, Liu H, Hu H, Grainger L, et al. Modulation of Fluorescent Protein Chromophores To Detect Protein Aggregation with Turn-On Fluorescence. J Am Chem Soc. 2018;140:7381-7384 pubmed publisher
  56. Meyer B, Segura J, Martinez K, Hovius R, George N, Johnsson K, et al. FRET imaging reveals that functional neurokinin-1 receptors are monomeric and reside in membrane microdomains of live cells. Proc Natl Acad Sci U S A. 2006;103:2138-43 pubmed
  57. Maurel D, Comps Agrar L, Brock C, Rives M, Bourrier E, Ayoub M, et al. Cell-surface protein-protein interaction analysis with time-resolved FRET and snap-tag technologies: application to GPCR oligomerization. Nat Methods. 2008;5:561-7 pubmed publisher
  58. Vauthier V, Derviaux C, Douayry N, Roux T, Trinquet E, Jockers R, et al. Design and validation of a homogeneous time-resolved fluorescence-based leptin receptor binding assay. Anal Biochem. 2013;436:1-9 pubmed publisher
  59. Clement C, Vassias I, Ray Gallet D, Almouzni G. Functional Characterization of Histone Chaperones Using SNAP-Tag-Based Imaging to Assess De Novo Histone Deposition. Methods Enzymol. 2016;573:97-117 pubmed publisher
  60. Keij J, BELL PRINCE C, Steinkamp J. Staining of mitochondrial membranes with 10-nonyl acridine orange, MitoFluor Green, and MitoTracker Green is affected by mitochondrial membrane potential altering drugs. Cytometry. 2000;39:203-10 pubmed
  61. Herb M, Gluschko A, Wiegmann K, Farid A, Wolf A, Utermöhlen O, et al. Mitochondrial reactive oxygen species enable proinflammatory signaling through disulfide linkage of NEMO. Sci Signal. 2019;12: pubmed publisher
  62. Schaupp L, Muth S, Rogell L, Kofoed Branzk M, Melchior F, Lienenklaus S, et al. Microbiota-Induced Type I Interferons Instruct a Poised Basal State of Dendritic Cells. Cell. 2020;181:1080-1096.e19 pubmed publisher
  63. Banik S, Pedram K, Wisnovsky S, Ahn G, Riley N, Bertozzi C. Lysosome-targeting chimaeras for degradation of extracellular proteins. Nature. 2020;584:291-297 pubmed publisher
  64. Zhang D, Liu J, Yin H, Wang H, Li S, Wang M, et al. A Turn on ESIPT Probe for Rapid and Ratiometric Fluorogenic Detection of Hg(2+) and its Application in Live-Cell Imaging. J Fluoresc. 2016;26:1367-72 pubmed publisher
  65. Anand T, Sivaraman G, Mahesh A, Chellappa D. Aminoquinoline based highly sensitive fluorescent sensor for lead(II) and aluminum(III) and its application in live cell imaging. Anal Chim Acta. 2015;853:596-601 pubmed publisher
  66. Goswamia S et al A colorimetric and ratiometric fluorescent turn-on fluoride chemodosimeter and application in live cell imaging: high selectivity via specific Si-O cleavage in semi-aqueous media and prompt recovery of ESIPT along with the X-ray structures. Tetrahedron Letters. 2014;55:2633–2638.
  67. Collot M, Fam T, Ashokkumar P, Faklaris O, Galli T, Danglot L, et al. Ultrabright and Fluorogenic Probes for Multicolor Imaging and Tracking of Lipid Droplets in Cells and Tissues. J Am Chem Soc. 2018;140:5401-5411 pubmed publisher
  68. Kazandjian T, Petras D, Robinson S, van Thiel J, Greene H, Arbuckle K, et al. Convergent evolution of pain-inducing defensive venom components in spitting cobras. Science. 2021;371:386-390 pubmed publisher
  69. Venkataramani V, Tanev D, Strahle C, Studier Fischer A, Fankhauser L, Kessler T, et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature. 2019;573:532-538 pubmed publisher
  70. Guo A, Wang Y, Chen B, Wang Y, Yuan J, Zhang L, et al. E-C coupling structural protein junctophilin-2 encodes a stress-adaptive transcription regulator. Science. 2018;362: pubmed publisher
  71. Zeng Q, Michael I, Zhang P, Saghafinia S, Knott G, Jiao W, et al. Synaptic proximity enables NMDAR signalling to promote brain metastasis. Nature. 2019;573:526-531 pubmed publisher
  72. Weber C, Zhou Y, Lee J, Looger L, Qian G, Ge C, et al. Temperature-dependent sex determination is mediated by pSTAT3 repression of Kdm6b. Science. 2020;368:303-306 pubmed publisher
  73. Helm P, Patwardhan A, Manders E. A study of the precision of confocal, ratiometric, Fura-2-based [Ca2+] measurements. Cell Calcium. 1997;22:287-98 pubmed
  74. Warner J, Hoshino A, Yamamoto K, Tilley R. Water-soluble photoluminescent silicon quantum dots. Angew Chem Int Ed Engl. 2005;44:4550-4 pubmed
  75. Alivisatos P. The use of nanocrystals in biological detection. Nat Biotechnol. 2004;22:47-52 pubmed
  76. E. Beaurepaire, V. Buissette, M.-P. Sauviat, D. Giaume, K. Lahlil, A. Mercuri, D. Casanova, A. Huignard, J.-L. Martin, T. Gacoin, J.-P. Boilot, and A. Alexandrou. Functionalized Fluorescent Oxide Nanoparticles: Artificial Toxins for Sodium Channel Targeting and Imaging at the Single- Molecule Level. Nano Lett., 4:2079–2083, 2004. Available from: dx.doi.org/10.1021/nl049105g
  77. Chang Y, Lee H, Chen K, Chang C, Tsai D, Fu C, et al. Mass production and dynamic imaging of fluorescent nanodiamonds. Nat Nanotechnol. 2008;3:284-8 pubmed publisher
  78. Faklaris O, Garrot D, Joshi V, Druon F, Boudou J, Sauvage T, et al. Detection of single photoluminescent diamond nanoparticles in cells and study of the internalization pathway. Small. 2008;4:2236-9 pubmed publisher
  79. Dahan M, Levi S, Luccardini C, Rostaing P, Riveau B, Triller A. Diffusion dynamics of glycine receptors revealed by single-quantum dot tracking. Science. 2003;302:442-5 pubmed
  80. Lee S, Jin C, Cai E, Ge P, Ishitsuka Y, Teng K, et al. Super-resolution imaging of synaptic and Extra-synaptic AMPA receptors with different-sized fluorescent probes. elife. 2017;6: pubmed publisher
  81. Kirchner C, Liedl T, Kudera S, Pellegrino T, Munoz Javier A, Gaub H, et al. Cytotoxicity of colloidal CdSe and CdSe/ZnS nanoparticles. Nano Lett. 2005;5:331-8 pubmed
  82. Lewinski N, Colvin V, Drezek R. Cytotoxicity of nanoparticles. Small. 2008;4:26-49 pubmed publisher
  83. Acharyya S, Oskarsson T, Vanharanta S, Malladi S, Kim J, Morris P, et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell. 2012;150:165-78 pubmed publisher
  84. Maguire C, Bovenberg M, Crommentuijn M, Niers J, Kerami M, Teng J, et al. Triple bioluminescence imaging for in vivo monitoring of cellular processes. Mol Ther Nucleic Acids. 2013;2:e99 pubmed publisher
  85. Evans M, Chaurette J, Adams S, Reddy G, Paley M, Aronin N, et al. A synthetic luciferin improves bioluminescence imaging in live mice. Nat Methods. 2014;11:393-5 pubmed publisher
  86. Mofford D, Reddy G, Miller S. Aminoluciferins extend firefly luciferase bioluminescence into the near-infrared and can be preferred substrates over D-luciferin. J Am Chem Soc. 2014;136:13277-82 pubmed publisher
  87. Saito K, Chang Y, Horikawa K, Hatsugai N, Higuchi Y, Hashida M, et al. Luminescent proteins for high-speed single-cell and whole-body imaging. Nat Commun. 2012;3:1262 pubmed publisher
  88. Suzuki K, Kimura T, Shinoda H, Bai G, Daniels M, Arai Y, et al. Five colour variants of bright luminescent protein for real-time multicolour bioimaging. Nat Commun. 2016;7:13718 pubmed publisher
  89. Chu J, Oh Y, Sens A, ATAIE N, Dana H, Macklin J, et al. A bright cyan-excitable orange fluorescent protein facilitates dual-emission microscopy and enhances bioluminescence imaging in vivo. Nat Biotechnol. 2016;34:760-7 pubmed publisher
  90. Yeh H, Karmach O, Ji A, Carter D, Martins Green M, Ai H. Red-shifted luciferase-luciferin pairs for enhanced bioluminescence imaging. Nat Methods. 2017;14:971-974 pubmed publisher
  91. Lukinavicius G, Blaukopf C, Pershagen E, Schena A, Reymond L, Derivery E, et al. SiR-Hoechst is a far-red DNA stain for live-cell nanoscopy. Nat Commun. 2015;6:8497 pubmed publisher
  92. Lukinavicius G, Reymond L, D Este E, Masharina A, Göttfert F, Ta H, et al. Fluorogenic probes for live-cell imaging of the cytoskeleton. Nat Methods. 2014;11:731-3 pubmed publisher
  93. Kilian N, Goryaynov A, Lessard M, Hooker G, Toomre D, Rothman J, et al. Assessing photodamage in live-cell STED microscopy. Nat Methods. 2018;15:755-756 pubmed publisher
  94. Wang H, Nakamura M, Abbott T, Zhao D, Luo K, Yu C, et al. CRISPR-mediated live imaging of genome editing and transcription. Science. 2019;: pubmed publisher
  95. Hafner A, Donlin Asp P, Leitch B, Herzog E, Schuman E. Local protein synthesis is a ubiquitous feature of neuronal pre- and postsynaptic compartments. Science. 2019;364: pubmed publisher
  96. Bink K, Walch A, Feuchtinger A, Eisenmann H, Hutzler P, Hofler H, et al. TO-PRO-3 is an optimal fluorescent dye for nuclear counterstaining in dual-colour FISH on paraffin sections. Histochem Cell Biol. 2001;115:293-9 pubmed
  97. Wang Q, Gao H, Clark K, Mugisha C, Davis K, Tang J, et al. CARD8 is an inflammasome sensor for HIV-1 protease activity. Science. 2021;371: pubmed publisher
  98. Nam S, Gupta V, Lee H, Lee J, Wisdom K, Varma S, et al. Cell cycle progression in confining microenvironments is regulated by a growth-responsive TRPV4-PI3K/Akt-p27Kip1 signaling axis. Sci Adv. 2019;5:eaaw6171 pubmed publisher
  99. Bon P, Lécart S, Fort E, Lévêque Fort S. Fast label-free cytoskeletal network imaging in living mammalian cells. Biophys J. 2014;106:1588-95 pubmed publisher
  100. Allen R, David G, Nomarski G. The zeiss-Nomarski differential interference equipment for transmitted-light microscopy. Z Wiss Mikrosk. 1969;69:193-221 pubmed
  101. Paddock S. Confocal laser scanning microscopy. Biotechniques. 1999;27:992-6, 998-1002, 1004 pubmed
  102. Stehbens S, Pemble H, Murrow L, Wittmann T. Imaging intracellular protein dynamics by spinning disk confocal microscopy. Methods Enzymol. 2012;504:293-313 pubmed publisher
  103. Yasuda S, Tsuchiya H, Kaiho A, Guo Q, Ikeuchi K, Endo A, et al. Stress- and ubiquitylation-dependent phase separation of the proteasome. Nature. 2020;578:296-300 pubmed publisher
  104. Magupalli V, Negro R, Tian Y, Hauenstein A, Di Caprio G, Skillern W, et al. HDAC6 mediates an aggresome-like mechanism for NLRP3 and pyrin inflammasome activation. Science. 2020;369: pubmed publisher
  105. Denk W, Strickler J, Webb W. Two-photon laser scanning fluorescence microscopy. Science. 1990;248:73-6 pubmed
  106. Millis B. Evanescent-wave field imaging: an introduction to total internal reflection fluorescence microscopy. Methods Mol Biol. 2012;823:295-309 pubmed publisher
  107. Reck Peterson S, Derr N, Stuurman N. Imaging single molecules using total internal reflection fluorescence microscopy (TIRFM). Cold Spring Harb Protoc. 2010;2010:pdb.top73 pubmed publisher
  108. Burchfield J, Lopez J, Mele K, Vallotton P, Hughes W. Exocytotic vesicle behaviour assessed by total internal reflection fluorescence microscopy. Traffic. 2010;11:429-39 pubmed publisher
  109. Calebiro D, Rieken F, Wagner J, Sungkaworn T, Zabel U, Borzi A, et al. Single-molecule analysis of fluorescently labeled G-protein-coupled receptors reveals complexes with distinct dynamics and organization. Proc Natl Acad Sci U S A. 2013;110:743-8 pubmed publisher
  110. Foerster T (1948) Zwischenmolekulare Energiewanderung und Fluoreszenz. Annalen des Physik (Leipzig) 2:55–75.
  111. Nakai J, Ohkura M, Imoto K. A high signal-to-noise Ca(2+) probe composed of a single green fluorescent protein. Nat Biotechnol. 2001;19:137-41 pubmed
  112. Mahajan N, Harrison Shostak D, Michaux J, Herman B. Novel mutant green fluorescent protein protease substrates reveal the activation of specific caspases during apoptosis. Chem Biol. 1999;6:401-9 pubmed
  113. DiPilato L, Cheng X, Zhang J. Fluorescent indicators of cAMP and Epac activation reveal differential dynamics of cAMP signaling within discrete subcellular compartments. Proc Natl Acad Sci U S A. 2004;101:16513-8 pubmed
  114. Ting A, Kain K, Klemke R, Tsien R. Genetically encoded fluorescent reporters of protein tyrosine kinase activities in living cells. Proc Natl Acad Sci U S A. 2001;98:15003-8 pubmed
  115. Wallrabe H, Periasamy A. Imaging protein molecules using FRET and FLIM microscopy. Curr Opin Biotechnol. 2005;16:19-27 pubmed
  116. Pelet S, Previte M, So P. Comparing the quantification of Forster resonance energy transfer measurement accuracies based on intensity, spectral, and lifetime imaging. J Biomed Opt. 2006;11:34017 pubmed
  117. Pfleger K, Eidne K. Illuminating insights into protein-protein interactions using bioluminescence resonance energy transfer (BRET). Nat Methods. 2006;3:165-74 pubmed
  118. Pfleger K, Seeber R, Eidne K. Bioluminescence resonance energy transfer (BRET) for the real-time detection of protein-protein interactions. Nat Protoc. 2006;1:337-45 pubmed
  119. Kocan M, See H, Seeber R, Eidne K, Pfleger K. Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cells. J Biomol Screen. 2008;13:888-98 pubmed publisher
  120. Breton B, Sauvageau E, Zhou J, Bonin H, Le Gouill C, Bouvier M. Multiplexing of multicolor bioluminescence resonance energy transfer. Biophys J. 2010;99:4037-46 pubmed publisher
  121. Goyet E, Bouquier N, Ollendorff V, Perroy J. Fast and high resolution single-cell BRET imaging. Sci Rep. 2016;6:28231 pubmed publisher
  122. Reits E, Neefjes J. From fixed to FRAP: measuring protein mobility and activity in living cells. Nat Cell Biol. 2001;3:E145-7 pubmed
  123. Xie H, Laird D, Chang T, Hu V. A mitosis-specific phosphorylation of the gap junction protein connexin43 in human vascular cells: biochemical characterization and localization. J Cell Biol. 1997;137:203-10 pubmed
  124. Köster M, Frahm T, Hauser H. Nucleocytoplasmic shuttling revealed by FRAP and FLIP technologies. Curr Opin Biotechnol. 2005;16:28-34 pubmed
  125. Van Craenenbroeck E, Engelborghs Y. Fluorescence correlation spectroscopy: molecular recognition at the single molecule level. J Mol Recognit. 2000;13:93-100 pubmed
  126. Mortensen K, Churchman L, Spudich J, Flyvbjerg H. Optimized localization analysis for single-molecule tracking and super-resolution microscopy. Nat Methods. 2010;7:377-81 pubmed publisher
  127. Kural C, Kim H, Syed S, Goshima G, Gelfand V, Selvin P. Kinesin and dynein move a peroxisome in vivo: a tug-of-war or coordinated movement?. Science. 2005;308:1469-72 pubmed
  128. Pack C, Yukii H, Toh e A, Kudo T, Tsuchiya H, Kaiho A, et al. Quantitative live-cell imaging reveals spatio-temporal dynamics and cytoplasmic assembly of the 26S proteasome. Nat Commun. 2014;5:3396 pubmed publisher
  129. Betzig E, Patterson G, Sougrat R, Lindwasser O, Olenych S, Bonifacino J, et al. Imaging intracellular fluorescent proteins at nanometer resolution. Science. 2006;313:1642-5 pubmed
  130. Wu Y, Wawrzusin P, Senseney J, Fischer R, Christensen R, Santella A, et al. Spatially isotropic four-dimensional imaging with dual-view plane illumination microscopy. Nat Biotechnol. 2013;31:1032-8 pubmed publisher
  131. Li D, Betzig E. Response to Comment on "Extended-resolution structured illumination imaging of endocytic and cytoskeletal dynamics". Science. 2016;352:527 pubmed publisher
  132. Serra D, Mayr U, Boni A, Lukonin I, Rempfler M, Challet Meylan L, et al. Self-organization and symmetry breaking in intestinal organoid development. Nature. 2019;569:66-72 pubmed publisher
  133. Rittscher J. Characterization of biological processes through automated image analysis. Annu Rev Biomed Eng. 2010;12:315-44 pubmed publisher
  134. Collins T. ImageJ for microscopy. Biotechniques. 2007;43:25-30 pubmed
  135. Yoo T, Ackerman M, Lorensen W, Schroeder W, Chalana V, Aylward S, et al. Engineering and algorithm design for an image processing Api: a technical report on ITK--the Insight Toolkit. Stud Health Technol Inform. 2002;85:586-92 pubmed
  136. Cromey D. Avoiding twisted pixels: ethical guidelines for the appropriate use and manipulation of scientific digital images. Sci Eng Ethics. 2010;16:639-67 pubmed publisher
ISSN : 2329-5139