Hostname: page-component-8448b6f56d-t5pn6 Total loading time: 0 Render date: 2024-04-23T09:47:24.066Z Has data issue: false hasContentIssue false

Focus on biosensors: Looking through the lens of quantitative biology

Published online by Cambridge University Press:  12 October 2021

James H. Rowe
Affiliation:
Sainsbury Laboratory, Cambridge University, Cambridge, United Kingdom
Alexander M. Jones*
Affiliation:
Sainsbury Laboratory, Cambridge University, Cambridge, United Kingdom
*
Author for correspondence: Alexander M. Jones, E-mail: alexander.jones@slcu.cam.ac.uk

Abstract

In recent years, plant biologists interested in quantifying molecules and molecular events in vivo have started to complement reporter systems with genetically encoded fluorescent biosensors (GEFBs) that directly sense an analyte. Such biosensors can allow measurements at the level of individual cells and over time. This information is proving valuable to mathematical modellers interested in representing biological phenomena in silico, because improved measurements can guide improved model construction and model parametrisation. Advances in synthetic biology have accelerated the pace of biosensor development, and the simultaneous expression of spectrally compatible biosensors now allows quantification of multiple nodes in signalling networks. For biosensors that directly respond to stimuli, targeting to specific cellular compartments allows the observation of differential accumulation of analytes in distinct organelles, bringing insights to reactive oxygen species/calcium signalling and photosynthesis research. In conjunction with improved image analysis methods, advances in biosensor imaging can help close the loop between experimentation and mathematical modelling.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2021. Published by Cambridge University Press in association with The John Innes Centre

1. The first glimmers of green light

Since the cryptic intron was removed from Aequorea victoria green fluorescent protein (GFP; Haseloff et al., Reference Haseloff, Siemering, Prasher and Hodge1997), fluorescent protein (FP) expression has had an incalculable impact on plant biology. The diverse uses of FPs include characterising protein expression pattern and localisation (Benková et al., Reference Benková, Michniewicz, Sauer, Teichmann, Seifertová, Jürgens and Friml2003), marking the endomembrane system (Geldner et al., Reference Geldner, Dénervaud-Tendon, Hyman, Mayer, Stierhof and Chory2009), demonstrating intracellular transcription factor movement (Nakajima et al., Reference Nakajima, Sena, Nawy and Benfey2001; Raissig et al., Reference Raissig, Matos, Gil, Kornfeld, Bettadapur, Abrash, Allison, Badgley, Vogel, Berry and Bergmann2017) and quantifying phloem unloading (Stadler et al., Reference Stadler, Wright, Lauterbach, Amon, Gahrtz, Feuerstein, Oparka and Sauer2004). Some of the most innovative FP uses have come from the development of GEFBs. These are proteins that contain one or more FPs, the fluorescent properties of which change with a given stimulus. This outlook piece will not detail the many biosensors used in plants [for a comprehensive overview, see a recent review (Walia et al., Reference Walia, Waadt and Jones2018)] but instead will emphasise recent developments, particularly of GEFBs that report on molecules and molecular events via direct sensing and independently of other components. We will then explore how biosensors are leading to a deeper more quantitative analysis of plant biology by providing more robust data for model construction and parametrisation.

2. From transcriptional reporters to direct biosensors

Over the years, FP expression has been placed under the control of a variety of promoters, many of which were chosen for their responsiveness to a given stimulus, such as a hormone. This principle was taken further with the development of synthetic promoters, which act as indirect hormone reporters, such as the auxin responsive DR5 promoter. In DR5::GFP, tandem repeats of an auxin responsive element are placed upstream of a minimal 35S Cauliflower Mosaic Virus promoter, to drive expression of GFP (Figure 1a; Benková et al., Reference Benková, Michniewicz, Sauer, Teichmann, Seifertová, Jürgens and Friml2003; Ulmasov et al., Reference Ulmasov, Murfett, Hagen and Guilfoyle1997). Higher FP fluorescence in a cell or tissue indicates sites of increased auxin response. Using fluorescent transcriptional reporters to infer hormone accumulation revolutionised plant developmental biology and acted as the basis for a series of spatial models of hormone distributions, helping establish the developmental regulation of patterning by auxin in both shoots and roots (Grieneisen et al., Reference Grieneisen, Xu, Marée, Hogeweg and Scheres2007; Jönsson et al., Reference Jönsson, Heisler, Shapiro, Meyerowitz and Mjolsness2006).

Fig. 1. Different types of genetically encoded fluorescent biosensors and their molecular mechanisms. (a) The mechanism of action of the auxin reporter DR5::GFP, where the auxin receptor complex targets the AUX/IAA transcriptional repressors for degradation, allowing green fluorescent protein transcription and fluorescence in the presence of the hormone. (b) The mechanism of action of the auxin biosensor DII:VENUS, where a venus fluorescent protein (FP) fused to domain II (DII) from an AUX/IAA protein is targeted for degradation by the auxin receptor complex, causing loss of fluorescence in the presence of the hormone. (c) The mechanism of action of the direct abscisic acid biosensor ABACUS1, where hormone binding causes the interaction of two sensory domains of a single fusion protein. This conformational change causes increased Förster resonance energy transfer (FRET) between two FP domains (a FRET donor and acceptor) and an altered emission ratio upon donor excitation. (d) The mechanism of action of the direct glutathione redox potential biosensor roGFP. A disulphide bond on the barrel of roGFP is reversibly sensitive to oxidation, causing an altered absorption spectrum.

This approach has been extremely useful and has consequently been used to create reporters for other hormones such as cytokinin and abscisic acid (ABA; Wu et al., Reference Wu, Duan, Pruneda-Paz, Oh, Pound, Kay and Dinneny2018; Zürcher et al., Reference Zürcher, Tavor-Deslex, Lituiev, Enkerli, Tarr and Müller2013), but at best transcriptional reporters such as DR5 indicate where hormone responses were. The levels and distribution of plant hormones are dynamically adjusted, to regulate a variety of processes such as tropisms (Band et al., Reference Band, Wells, Fozard, Ghetiu, French, Pound, Wilson, Yu, Li, Hijazi, Oh, Pearce, Perez-Amador, Yun, Kramer, Alonso, Godin, Vernoux, Hodgman and Bennett2014), developmental patterning (Benková et al., Reference Benková, Michniewicz, Sauer, Teichmann, Seifertová, Jürgens and Friml2003) and stress responses (Iuchi et al., Reference Iuchi, Kobayashi, Yamaguchi-Shinozaki and Shinozaki2000; Rowe et al., Reference Rowe, Topping, Liu and Lindsey2016). The time required for transcription, translation and maturation of FPs (Balleza et al., Reference Balleza, Kim and Cluzel2018) mean that fluorescence increases following stimuli are delayed; in fact, a DR5::VENUS auxin response does not become visible until 2 hr after the mRNA accumulation and follows different dynamics (Brunoud et al., Reference Brunoud, Wells, Oliva, Larrieu, Mirabet, Burrow, Beeckman, Kepinski, Traas, Bennett and Vernoux2012). FP longevity is such that a reduction in fluorescence following removal of a stimulus could be even slower, for example, wildtype GFP has a 26-hr half-life in mammalian systems (Corish & Tyler-Smith, Reference Corish and Tyler-Smith1999). Transcriptional reporters are indirect and so rely on endogenous signalling components and transcriptional/translational machinery, which may vary over time or from tissue to tissue (Bargmann et al., Reference Bargmann, Vanneste, Krouk, Nawy, Efroni, Shani, Choe, Friml, Bergmann, Estelle and Birnbaum2013; Prigge et al., Reference Prigge, Platre, Kadakia, Zhang, Greenham, Szutu, Pandey, Bhosale, Bennett, Busch and Estelle2020).

The next big advance in imaging hormone responses came with the advent of degradation-based reporters such as DII-VENUS. The second domain (DII) from IAA28 was fused to the yellow FP Venus, to create a fluorescent fusion protein that is ubiquitinated by the SCFTIR1 complex upon auxin perception (Figure 1b; Brunoud et al., Reference Brunoud, Wells, Oliva, Larrieu, Mirabet, Burrow, Beeckman, Kepinski, Traas, Bennett and Vernoux2012). DII-VENUS ubiquitination causes rapid degradation and loss of fluorescence, reducing the time delay between hormone accumulation and response (Brunoud et al., Reference Brunoud, Wells, Oliva, Larrieu, Mirabet, Burrow, Beeckman, Kepinski, Traas, Bennett and Vernoux2012). This facilitated the modelling of fast root auxin redistribution due to gravistimulation (Band et al., Reference Band, Wells, Fozard, Ghetiu, French, Pound, Wilson, Yu, Li, Hijazi, Oh, Pearce, Perez-Amador, Yun, Kramer, Alonso, Godin, Vernoux, Hodgman and Bennett2014). Whilst this is a great improvement in imaging hormone accumulations, problems arise in determining whether differences in fluorescence are due to changes in expression or degradation of the FP. A powerful tool to overcome this problem is the expression of a second FP, as in the R2D2 or qDII reporters (Galvan-Ampudia et al., Reference Galvan-Ampudia, Cerutti, Legrand, Brunoud, Martin-Arevalillo, Azais, Bayle, Moussu, Wenzl, Jaillais, Lohmann, Godin and Vernoux2020; Liao et al., Reference Liao, Smet, Brunoud, Yoshida, Vernoux and Weijers2015). By comparing the DII-VENUS fluorescence, which is lost in the presence of auxin, to a stabilised control FP, tissue specific expression and optical differences are taken into account, leading to better estimates of auxin concentration. However, DII-VENUS, R2D2 and qDII still rely on the endogenous SCFTIR1 and proteasomal machinery to elicit fluorescence changes, so could experience tissue to tissue variation in response, and they cannot be used to image fast hormone depletions or oscillations as VENUS maturation can take upwards of 18 min in bacterial systems (Balleza et al., Reference Balleza, Kim and Cluzel2018).

One approach to overcome these problems is to engineer a fluorescent biosensor that changes its fluorescence properties depending on direct interaction with the molecule or molecular event of interest and independently of other components; these are the direct GEFBs. Many also have a ratiometric output to control for optical artefacts (such as the loss of signal in deeper tissues) during imaging and differences in expression.

Whilst direct GEFBs for small second messengers, such as calcium, have existed for a long time (Miyawaki et al., Reference Miyawaki, Llopis, Heim, McCaffery, Adams, Ikura and Tsien1997), the first direct plant hormone GEFBs were developed fairly recently and all follow a similar design principle to previous ratiometric Förster resonance energy transfer (FRET) sensors (Herud-Sikimić et al., Reference Herud-Sikimic, Stiel, Kolb, Shanmugaratnam, Berendzen, Feldhaus, Höcker and Jürgens2021; Jones et al., Reference Jones, Danielson, ManojKumar, Lanquar, Grossmann and Frommer2014; Rizza et al., Reference Rizza, Walia, Lanquar, Frommer and Jones2017; Waadt et al., Reference Waadt, Hitomi, Nishimura, Hitomi, Adams, Getzoff and Schroeder2014). The abscisic acid concentration and uptake sensor 1-2μ (ABACUS1-2μ), for example, consists of a sensory domain and two FPs, all connected by linkers to form a single protein (Figure 1c). The FPs undergo FRET from the donor edCerulean to the acceptor edCitrine upon donor excitation. The amount of energy transfer is sensitive to the distance and orientation of the FPs (Jares-Erijman & Jovin, Reference Jares-Erijman and Jovin2003). Upon ABA binding, the biosensor undergoes a conformational change, which causes changes in FRET and thereby in FP emission ratio (Jones et al., Reference Jones, Danielson, ManojKumar, Lanquar, Grossmann and Frommer2014). ABACUS1-2μ is an example of an extrinsic sensor, as it relies on a sensory domain separate to the fluorescence domain to respond to a stimulus.

Intrinsic sensors, where the fluorescence properties of the FP itself are varied by direct interactions with the stimulus, also exist (Walia et al., Reference Walia, Waadt and Jones2018). roGFP, for example, is an intrinsic ratiometric sensor for the glutathione redox potential (EGSH; Figure 1d; Hanson et al., Reference Hanson, Aggeler, Oglesbee, Cannon, Capaldi, Tsien and Remington2004). A disulphide bond on the barrel of roGFP is reversibly sensitive to oxidation, causing an altered absorption spectrum. By sequentially exciting the FP at two different wavelengths, and quantifying emission, a ratio of the oxidised and reduced roGFP is obtained, which indicates EGSH at the subcellular level (Hanson et al., Reference Hanson, Aggeler, Oglesbee, Cannon, Capaldi, Tsien and Remington2004; Jiang et al., Reference Jiang, Schwarzer, Lally and Feldman2006).

There are too many other sensor types and designs to be summarised here, and sensor development is the subject of much ongoing research (Walia et al., Reference Walia, Waadt and Jones2018). An ideal sensor is reversible, ratiometric, specific, responsive to the analyte in the endogenous range, has minimal impact on endogenous signalling (i.e., highly orthogonal) and therefore minimal phenotypes and has a large signal-to-noise ratio. Developing sensors that fulfil all of these criteria can take decades, but even imperfect sensors can be useful in a host of research contexts. As numerous sensors for diverse analytes are now widely available, researchers can simultaneously look at different steps in complex systems to quantitatively understand signal processing at high spatiotemporal resolution as detailed in the next section.

3. Sensing signalling in time and space

A biological system will often contain interacting multireaction pathways, which vary the concentration of a host of chemical species. When representing a system in a model, choosing which interactions to include and exclude is a difficult process that strongly affects the model behaviour and predictions (Aldridge et al., Reference Aldridge, Burke, Lauffenburger and Sorger2006). Models are often simplified for a lack of information or for computational reasons, as reducing the number of unconstrained parameters vastly reduces the computation required for parameter scanning. A common simplification is to reduce a multistep signalling pathway to single reaction, with the assumption that this will not affect the model outcomes, but recent work has shown that this often sacrifices the ability of the model to reproduce temporal dynamics (Korsbo & Jönsson, Reference Korsbo and Jönsson2020). Using transcriptional reporter systems, the experimental outputs are abstracted from the signalling by multiple steps, so the temporal dynamics have been difficult to measure, making model validation difficult.

But for many signalling networks, there are now direct GEFBs for multiple steps, which can be tracked in real time in living cells, allowing the network to be dissected in fine detail. This is exemplified in abiotic stress responses which often involve responses in calcium, ABA, Sucrose nonfermenting-1-related protein kinase 2 activity and reactive oxygen species, which may show differing yet overlapping dynamics to regulate processes such as gene expression and stomatal closure. Each of these signalling steps now also has a suite of direct biosensors (Huang et al., Reference Huang, Waadt, Nuhkat, Kollist, Hedrich and Roelfsema2019; Jones et al., Reference Jones, Danielson, ManojKumar, Lanquar, Grossmann and Frommer2014; Waadt et al., Reference Waadt, Hitomi, Nishimura, Hitomi, Adams, Getzoff and Schroeder2014; Zhang et al., Reference Zhang, Takahashi, Hsu, Kollist, Merilo, Krysan and Schroeder2020), allowing high spatiotemporal quantification.

Recent pioneering co-expression of pairs of direct biosensors has even allowed the temporal dynamics of many second messengers or hormones to be characterised simultaneously (ABA and Ca2+; pH and EGSH; pH and H2O2; Ca2+ and EGSH; Ca2+ and H2O2; Ca2+, pH and Cl (Keinath et al., Reference Keinath, Waadt, Brugman, Schroeder, Grossmann, Schumacher and Krebs2015; Waadt et al., Reference Waadt, Krebs, Kudla and Schumacher2017; Reference Waadt, Köster, Andrés, Waadt, Bradamante, Lampou, Kudla and Schumacher2020). This has confirmed many predictions and offered new insights. Whereas auxin application caused the fast modulation Ca2+ and H+ dynamics in roots, ABA application caused no such fast ion dynamics. This response stands in contrast with stomata, where ABA microinjection elicits a Ca2+ response in the majority of stomata, enhancing closure (Huang et al., Reference Huang, Waadt, Nuhkat, Kollist, Hedrich and Roelfsema2019; Waadt et al., Reference Waadt, Krebs, Kudla and Schumacher2017). Such detailed datasets will no doubt be invaluable in constructing realistic models of signal integration in the future.

In addition to temporal responses, spatial signalling responses can also be measured with biosensors. Unlike transcriptional/degron-based reporters, direct GEFBs that independently sense their molecule or molecular event of interest allow the subcellular dynamics of signalling systems to be assessed. This has revealed the spatial importance of subcellular localisation of calcium accumulation in root growth (Leitão et al., Reference Leitão, Dangeville, Carter and Charpentier2019) and the differing redox status of different organelles (Exposito-Rodriguez et al., Reference Exposito-Rodriguez, Laissue, Yvon-Durocher, Smirnoff and Mullineaux2017; Schwarzländer et al., Reference Schwarzländer, Fricker, Müller, Marty, Brach, Novak, Sweetlove, Hell and Meyer2008). Most biosensors so far have focused on the nuclear or cytoplasmic space, but as experimentalists are targeting biosensors to other cellular compartments, so we will see a whole host of new, spatially distinct responses in the coming years, allowing the modelling and understanding of interorganelle signalling.

Unfortunately, many biosensors currently in use contain FPs that might be unsuitable for highly oxidising or acidic compartments present in some organelles or the apoplasm, although changing the FPs may help address this (Erard & Guiot, Reference Erard and Guiot2015). The FP engineering community has constantly strived to produce a diverse pallet of FPs that are now listed in FPbase, providing an invaluable tool for biosensor engineering. FPbase is a repository, which consolidates the fluorescence spectra, quantum yield, acid sensitivity (i.e., pKa), key mutations and other important properties for a vast number of FPs in a searchable format (Lambert, Reference Lambert2019). When combined with increasingly affordable nucleic acid synthesis technology, high throughput combinatorial cloning techniques provide a powerful platform for rapid biosensor engineering. The expanding diversity of GEFBs and their applications has necessitated improved analysis methods to extract and interpret diverse datasets.

4. Image analysis and data processing, overcoming a major bottleneck to interpreting large datasets

In recent years, there has been a renaissance in development of imaging modalities and analysis methods. For analysis, simple tools like ImageJ have been expanded into flexible toolsets like Fiji (Fiji Is Just ImageJ), with multitudes of user-written plug-ins (Schindelin et al., Reference Schneider, Rasband and Eliceiri2012), and the use of comprehensive analysis suites, such as Morphographx/LithographX (de Reuille et al., Reference de Reuille, Routier-Kierzkowska, Kierzkowski, Bassel, Schüpbach, Tauriello, Bajpai, Strauss, Weber, Kiss, Burian, Hofhuis, Sapala, Lipowczan, Heimlicher, Robinson, Bayer, Basler, Koumoutsakos and Smith2015) or IMARIS (https://imaris.oxinst.com/), is now commonplace, offering compelling nearly complete segmentations and reconstructions of entire organs.

The first step in image analysis is often segmentation (Figure 2), the process of deciding which parts of an image are of interest for analysis and labelling them so data can be extracted (Long et al., Reference Long, Zhou and Peng2012). At the simplest level, this involves user-drawn regions of interest (ROIs) in tools like ImageJ, whilst more complex analysis may involve automated/semi-automated segmentation performed computationally.

Fig. 2. Example image processing pipeline for the nuclear localised ratiometric Förster resonance energy transfer sensor nlsABACUS1-2μ. Notes: Several image acquisition channels are used in FRET biosensor analysis. Here, the new FRETENATOR analysis pipeline for Fiji was used (Rowe et al., Reference Rowe, Rizza and Jones2021). For segmentation, the following steps are performed: first a difference of Gaussian filter is applied to the acceptor-excited acceptor-emission channel to remove background and smooth noise, then Otsu’s method is used to threshold the filtered image based on signal intensity. A watershed algorithm is used to split touching objects in the threshold image, and a connected-component analysis is used to label all the individual objects, producing a label map. The quantification steps involve removing saturated voxels from the original images and then dividing the mean signal intensity of donor-excited acceptor-emission channel by the donor-exited donor-emission channel for each labelled ROI, which can be represented on the segmented image using false coloration (e.g., the Turbo lookup table used here).

The importance of a good segmentation becomes apparent in the subsequent quantification steps. For intensiometric sensors, this involves quantifying one signal channel, whereas for ratiometric sensors, this may involve dividing the signal from one channel by another (Mahlandt & Goedhart, Reference Mahlandt and Goedhart2021; Rizza et al., Reference Rizza, Walia, Tang and Jones2019). For ratiometric sensors, quantifying fluorescence emission in areas with poor/no signal can lead to artefactually large variation and a low signal-to-noise ratio as can including pixels where the detector is saturated—such areas should be removed prior to interpretation (Figure 2).

Focal drift, sample movement and growth also present problems for time course imaging and analysis. Often depth (z-plane) movement causes brightness changes (an optical artefact), which is a particular problem for intensiometric GEFBs. During analysis, if ROIs fail to move in step with the moving object, then additional artefacts may be introduced. Imaging samples in microfluidics systems to limit sample movement can help eliminate some of these issues, but often they are dealt with during the analysis stage. One solution is to identify features that move and offset each timeframe in an image sequence to eliminate movement (image registration and translation). If object displacement is not uniform, such as in a growing tissue, then tracking of segmented objects may help address any artefacts from movement.

Biosensor images present further challenges for analysis, as they often require multichannel imaging and sequential acquisition, which must be taken at high speed when measuring fast dynamics. Because of the complexity of the imaging setup, a compromise is often made between acquisition speed, resolution and signal-to-noise ratio (Rizza et al., Reference Rizza, Walia, Tang and Jones2019; Rowe et al., Reference Rowe, Rizza and Jones2021). This compromise means accurate fully automated segmentation and complete tissue reconstruction is difficult, but the development of deep learning segmentation tools, such as PlantSeg (Wolny et al., Reference Wolny, Cerrone, Vijayan, Tofanelli, Barro, Louveaux, Wenzl, Strauss, Wilson-Sánchez, Lymbouridou, Steigleder, Pape, Bailoni, Duran-Nebreda, Bassel, Lohmann, Tsiantis, Hamprecht, Schneitz and Kreshuk2020) and Stardist (Weigert et al., Reference Weigert, Schmidt, Haase, Sugawara and Myers2020), may help increase segmentation accuracy and overcome some of these bottlenecks. After segmentation steps, biosensor image analysis workflows also require additional bespoke steps, such as background subtraction and the calculation of emission ratios, to process the data, so it can be quantified meaningfully (Mahlandt & Goedhart, Reference Mahlandt and Goedhart2021; Rizza et al., Reference Rizza, Walia, Lanquar, Frommer and Jones2017; Rowe et al., Reference Rowe, Rizza and Jones2021; Waadt et al., Reference Waadt, Köster, Andrés, Waadt, Bradamante, Lampou, Kudla and Schumacher2020).

Without automation, the image analysis process can become laborious and one of the main bottlenecks in biosensor labs, taking much longer than the imaging itself. This bottleneck can be overcome with flexible software environments, such as Fiji or Icy, where custom workflows can be made, tested and applied quickly in batch (De Chaumont et al., Reference De Chaumont, Dallongeville, Chenouard, Hervé, Pop, Provoost, Meas-Yedid, Pankajakshan, Lecomte, Le Montagner, Lagache, Dufour and Olivo-Marin2012; Schindelin et al., Reference Schneider, Rasband and Eliceiri2012).

Combining high-resolution biosensor image sets with fully labelled and segmented image analysis tools may soon be widely achievable with use of deep learning tools. This would offer many exciting possibilities for future work, for example, correlating the size, position and growth rates of all cells within an organ with hormone concentrations, or examining how cellular organisation and connections affect hormone accumulation sites. These sorts of approaches have already yielded impressive results with the transcriptional and degron reporters. Models of root auxin flux based on cell geometries and transporter levels extracted from images have demonstrated the importance of plasmodesmatal connections to reproduce measured DII-VENUS patterns (Mellor et al., Reference Mellor, Voß, Janes, Bennett, Wells and Band2020). As the integration of hormone responses with precise spatial information has allowed transport models to recapitulate auxin response patterns, direct GEFBs will spur integrated models of in planta hormone biochemical pathways to dissect complex phenomena such as hormone distributions.

5. Breaking down biochemistry… and building it up

Biochemical pathways can be examined in models by representing component reactions numerically. A challenge is that parameters determining reaction rates are often difficult to directly measure. Statistical approaches are usually employed to explore the parameter space and identify which parameters have the greatest influence on model behaviour (Aldridge et al., Reference Aldridge, Burke, Lauffenburger and Sorger2006; Vernon et al., Reference Vernon, Liu, Goldstein, Rowe, Topping and Lindsey2018). Parameterisation is hampered by the difficulty of measuring the concentrations of individual chemical species, leaving many parameters unconstrained.

Biosensors allow the real-time quantification of metabolites, in planta, which greatly improves the parametrisation process. By using knockouts, applying reaction precursors and using inducible genetics, the relative contributions of various steps in a pathway can now be quantified. This approach was recently taken by Rizza et al. (Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021), to characterise how a gibberellin gradient is established in the Arabidopsis root.

Previous multiscale modelling based on the expression of a Gibberellin 20-oxidase (GA20ox) biosynthesis gene had predicted that gibberellin levels would reach a maximum at the start of the root elongation zone and would diminish via dilution as cells elongated and progressed to the differentiation zone (Band et al., Reference Band, Úbeda-Tomás, Dyson, Middleton, Hodgman, Owen, Jensen, Bennett and King2012). This contrasted with the gibberellin distribution observed with the gibberellin sensitive nlsGPS1 FRET biosensor (Rizza et al., Reference Rizza, Walia, Lanquar, Frommer and Jones2017), which showed low gibberellin levels in the meristem that increased as the cells elongated. Therefore, Rizza et al. (Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021) performed iterative gibberellin measurements, perturbations and mathematical modelling to determine that a gradual increase in gibberellin biosynthesis across the elongation zone best explains how the gibberellin distribution is generated.

Rather than the presumed rate limiting GA20ox enzyme step, both early biosynthetic steps and the final Gibberellin 3-oxidase (GA3ox) catalysed step were locally rate limiting in the elongation zone, the site of high gibberellin accumulation in Arabidopsis roots (Rizza et al., Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021). In contrast, GA20ox along with GA3ox biosynthetic steps were together rate limiting for gibberellin accumulation in the most apical meristematic region. Exogenously applied gibberellin reinforced the endogenous gradient, rather than eliminating it, a behaviour that was best reproduced in silico by assuming differential plasma membrane permeability to gibberellin (Rizza et al., Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021). The model predicted that artificially increasing permeability along the root would eliminate the gibberellin gradient when exogenous hormone is applied, which was tested in vivo by briefly applying gibberellin at low exogenous pH where indeed the gradient was greatly reduced.

This level of resolution when dissecting the functionally relevant steps of a biochemical pathway across an organ was made more accessible through the use of direct GEFBs and inspires further questions. Although the nlsGPS1 biosensor illustrates a correlation between gibberellin and cellular growth (Rizza et al., Reference Rizza, Walia, Lanquar, Frommer and Jones2017), it is now clear from gibberellin perturbation experiments measuring gibberellin changes alongside growth that a simple dose-response model is insufficient to explain the relationship between the hormone and root cell behaviour (Rizza et al., Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021). This work demonstrates the power of the fast spatiotemporal responses of biosensors, in reliably quantifying metabolites and accurately parametrising biological models to move research forward quantitatively.

6. Focus on the future

Plant biosensors are already starting to move plant biology in a more quantitative direction. Complex metabolic processes are now being dissected at the level of spatially separate yet interconnected steps in vivo. A striking example is the recent characterisation of the photosynthesis dependent changes in NADP(H) and NAD(H) in chloroplasts, mitochondria, cytosol and peroxisomes using the iNAP and SoNar sensors (Lim et al., Reference Lim, Voon, Guan, Yang, Gardeström and Lim2020). Using elegant combinations of biosensors, inhibitors and illumination treatments, this work demonstrates the flow of reducing equivalents between chloroplasts, peroxisomes and mitochondria through the cytoplasm to support photorespiration. The compartmentalisation of different biochemical processes into cells and organelles is an essential part of eukaryotic life, but it has historically proved challenging to study the interactions of these primary metabolic pathways in situ.

Targeting biosensors to different tissues and compartments like this may sometimes require some sensor re-engineering, but it is an approach that is already yielding great new insights into signalling, revealing the spatial specificity of responses. The targeting of R-GECO1.2 and G-GECO1.2 to the cytoplasm and nucleus demonstrated nuclear calcium spikes in Arabidopsis (Kelner et al., Reference Kelner, Leitão, Chabaud, Charpentier and de Carvalho-Niebel2018; Leitão et al., Reference Leitão, Dangeville, Carter and Charpentier2019), and genetically disrupting these spikes with mutations in a nuclear cation transporter causes altered auxin homeostasis and root growth (Leitão et al., Reference Leitão, Dangeville, Carter and Charpentier2019).

Continued biosensor development may offer the tools to answer many of the big questions within plant biology, as new biosensors for difficult to measure metabolites, hormones, molecular events and even biophysical conditions are developed. Innovative new sensors, such as a recently developed crowding sensor for osmolarity (Cuevas-Velazquez et al., Reference Cuevas-Velazquez, Vellosillo, Guadalupe, Schmidt, Yu, Moses, Brophy, Cosio-Acosta, Das, Wang, Jones, Covarrubias, Sukenik and Dinneny2021), will allow the experimental integration of stress responses, biomechanics and gene expression.

The ongoing revolution in image analysis offers many exciting possibilities for future work with biosensors, through the correlation of spatial and tissue organisational data with biosensor outputs. High spatiotemporal resolution, nondestructive imaging in conjunction with comprehensive image analysis allows access to information that was unimaginable a few years ago. This approach was recently used to demonstrate a temporal centrifugal wave of auxin dynamics that precedes growth in the shoot apical meristem, by using degron-based auxin reporters (Galvan-Ampudia et al., Reference Galvan-Ampudia, Cerutti, Legrand, Brunoud, Martin-Arevalillo, Azais, Bayle, Moussu, Wenzl, Jaillais, Lohmann, Godin and Vernoux2020). Measuring hormone concentrations in situ has also improved models demonstrating which biochemical steps are essential to create and maintain hormone gradients (Rizza et al., Reference Rizza, Tang, Stanley, Grossmann, Owen, Band and Jones2021). These methods are broadly applicable, and the construction and parameterisation of a variety of biochemical and morphodynamic models will be improved with better data. This is perhaps the most exciting use of biosensor data—to close the loop between experimentation and mathematical modelling. The quantitative nature of biosensors offers many exciting future possibilities in plant research, and, though not yet measurable, the future seems bright.

Acknowledgements

The authors would like to thank Martin Balcerowicz and Marino Exposito-Rodriguez for their helpful comments on the manuscript.

Financial support

The authors would like to thank their funders: the Gatsby Charitable Foundation and BBSRC (BB/P018572/1).

Conflicts of interest

The authors would like to declare they have no conflicts of interest.

Authorship contributions

JHR and AMJ wrote the manuscript. JHR made the figures.

Data availability statement

Code for the analysis presented in Figure 2, as well as test dataset, is available at https://github.com/JimageJ/ImageJ-Tools.

References

Aldridge, B. B., Burke, J. M., Lauffenburger, D. A., & Sorger, P. K. (2006). Physicochemical modelling of cell signalling pathways. Nature Cell Biology, 8, 11951203.CrossRefGoogle ScholarPubMed
Balleza, E., Kim, J. M., & Cluzel, P. (2018). Systematic characterization of maturation time of fluorescent proteins in living cells. Nature Methods, 15, 4751.CrossRefGoogle ScholarPubMed
Band, L. R., Úbeda-Tomás, S., Dyson, R. J., Middleton, A. M., Hodgman, T. C., Owen, M. R., Jensen, O. E., Bennett, M. J., & King, J. R. (2012). Growth-induced hormone dilution can explain the dynamics of plant root cell elongation. Proceedings of the National Academy of Sciences of the United States of America, 109, 75777582. https://doi.org/10.1073/pnas.1113632109.CrossRefGoogle ScholarPubMed
Band, L. R., Wells, D. M., Fozard, J. A., Ghetiu, T., French, A. P., Pound, M. P., Wilson, M. H., Yu, L., Li, W., Hijazi, H. I., Oh, J., Pearce, S. P., Perez-Amador, M. A., Yun, J., Kramer, E., Alonso, J. M., Godin, C., Vernoux, T., Hodgman, T. C., … Bennett, M. J. (2014). Systems analysis of auxin transport in the Arabidopsis root apex. Plant Cell, 26, 862875. https://doi.org/10.1105/tpc.113.119495.CrossRefGoogle ScholarPubMed
Bargmann, B. O. R., Vanneste, S., Krouk, G., Nawy, T., Efroni, I., Shani, E., Choe, G., Friml, J., Bergmann, D. C., Estelle, M., & Birnbaum, K. D. (2013). A map of cell type‐specific auxin responses. Molecular Systems Biology, 9, 688. https://doi.org/10.1038/msb.2013.40.Google ScholarPubMed
Benková, E., Michniewicz, M., Sauer, M., Teichmann, T., Seifertová, D., & Jürgens, G., Friml, J. (2003). Local, efflux-dependent auxin gradients as a common module for plant organ formation. Cell, 115, 591602. https://doi.org/10.1016/S0092-8674(03)00924-3.Google ScholarPubMed
Brunoud, G., Wells, D. M., Oliva, M., Larrieu, A., Mirabet, V., Burrow, A. H., Beeckman, T., Kepinski, S., Traas, J., Bennett, M. J., & Vernoux, T. (2012). A novel sensor to map auxin response and distribution at high spatio-temporal resolution. Nature, 482 103106. https://doi.org/10.1038/nature10791.Google ScholarPubMed
Corish, P., & Tyler-Smith, C. (1999). Attenuation of green fluorescent protein half-life in mammalian cells. Protein Engineering, Design and Selection, 12, 10351040.CrossRefGoogle ScholarPubMed
Cuevas-Velazquez, C. L., Vellosillo, T., Guadalupe, K., Schmidt, B., Yu, F., Moses, D., Brophy, J. A., Cosio-Acosta, D., Das, A., Wang, L., Jones, A. M., Covarrubias, A. A., Sukenik, S., & Dinneny, J. R. (2021). Intrinsically disordered protein biosensor tracks the physical-chemical effects of osmotic stress on cells. BioRxiv. https://doi.org/10.1101/2021.02.17.431712.CrossRefGoogle Scholar
De Chaumont, F., Dallongeville, S., Chenouard, N., Hervé, N., Pop, S., Provoost, T., Meas-Yedid, V., Pankajakshan, P., Lecomte, T., Le Montagner, Y., Lagache, T., Dufour, A., & Olivo-Marin, J. C. (2012). Icy: An open bioimage informatics platform for extended reproducible research. Nature Methods, 9, 690696. https://doi.org/10.1038/nmeth.2075.CrossRefGoogle ScholarPubMed
de Reuille, P. B., Routier-Kierzkowska, A.-L. L., Kierzkowski, D., Bassel, G. W., Schüpbach, T., Tauriello, G., Bajpai, N., Strauss, S., Weber, A., Kiss, A., Burian, A., Hofhuis, H., Sapala, A., Lipowczan, M., Heimlicher, M. B., Robinson, S., Bayer, E. M., Basler, K., Koumoutsakos, P., … Smith, R. S. (2015). MorphoGraphX: A platform for quantifying morphogenesis in 4D. eLife, 4, e05864. https://doi.org/10.7554/eLife.05864.CrossRefGoogle Scholar
Erard, M., & Guiot, E. (2015). pH sensitivity of FRET reporters based on cyan and yellow fluorescent proteins. Analytical and Bioanalytical Chemistry, 407, 41834193. https://doi.org/10.1007/s00216-015-8636-z.Google Scholar
Exposito-Rodriguez, M., Laissue, P. P., Yvon-Durocher, G., Smirnoff, N., & Mullineaux, P. M. (2017). Photosynthesis-dependent H2O2 transfer from chloroplasts to nuclei provides a high-light signalling mechanism. Nature Communications, 8, 111.CrossRefGoogle ScholarPubMed
Galvan-Ampudia, C. S., Cerutti, G., Legrand, J., Brunoud, G., Martin-Arevalillo, R., Azais, R., Bayle, V., Moussu, S., Wenzl, C., Jaillais, Y., Lohmann, J. U., Godin, C., & Vernoux, T. (2020). Temporal integration of auxin information for the regulation of patterning. eLife, 9, 165. https://doi.org/10.7554/eLife.55832.CrossRefGoogle ScholarPubMed
Geldner, N., Dénervaud-Tendon, V., Hyman, D. L., Mayer, U., Stierhof, Y. D., & Chory, J. (2009). Rapid, combinatorial analysis of membrane compartments in intact plants with a multicolor marker set. Plant Journal, 59, 169178.CrossRefGoogle ScholarPubMed
Grieneisen, V. A., Xu, J., Marée, A. F. M., Hogeweg, P., & Scheres, B. (2007). Auxin transport is sufficient to generate a maximum and gradient guiding root growth. Nature, 449, 10081013.CrossRefGoogle ScholarPubMed
Hanson, G. T., Aggeler, R., Oglesbee, D., Cannon, M., Capaldi, R. A., Tsien, R. Y., & Remington, S. J. (2004). Investigating mitochondrial redox potential with redox-sensitive green fluorescent protein indicators. Journal of Biological Chemistry, 279, 1304413053. https://doi.org/10.1074/jbc.M312846200.Google ScholarPubMed
Haseloff, J., Siemering, K. R., Prasher, D. C., & Hodge, S. (1997). Removal of a cryptic intron and subcellular localization of green fluorescent protein are required to mark transgenic Arabidopsis plants brightly. Proceedings of the National Academy of Sciences of the United States of America, 94, 21222127.CrossRefGoogle ScholarPubMed
Herud-Sikimic, O., Stiel, A. C., Kolb, M., Shanmugaratnam, S., Berendzen, K. W., Feldhaus, C., Höcker, B., & Jürgens, G. (2021). A biosensor for the direct visualization of auxin. Nature, 592, 768772. https://doi.org/10.1038/s41586-021-03425-2.Google ScholarPubMed
Huang, S., Waadt, R., Nuhkat, M., Kollist, H., Hedrich, R., & Roelfsema, M. R. G. (2019). Calcium signals in guard cells enhance the efficiency by which abscisic acid triggers stomatal closure. New Phytologist, 224, 177187.Google ScholarPubMed
Iuchi, S., Kobayashi, M., Yamaguchi-Shinozaki, K., & Shinozaki, K. (2000). A stress-inducible gene for 9-cis-epoxycarotenoid dioxygenase involved in abscisic acid biosynthesis under water stress in drought-tolerant cowpea. Plant Physiology, 123, 553562.CrossRefGoogle ScholarPubMed
Jares-Erijman, E. A., & Jovin, T. M. (2003). FRET imaging. Nature biotechnology, 21, 13871395. https://doi.org/10.1038/nbt896.CrossRefGoogle ScholarPubMed
Jiang, K., Schwarzer, C., Lally, E., … Feldman, L. (2006). Expression and characterization of a redox-sensing green fluorescent protein (reduction-oxidation-sensitive green fluorescent protein) in Arabidopsis. Plant Physiology, 141, 397403.CrossRefGoogle ScholarPubMed
Jones, A. M., Danielson, J. Å. H., ManojKumar, S. N., Lanquar, V., Grossmann, G., & Frommer, W. B. (2014). Abscisic acid dynamics in roots detected with genetically encoded FRET sensors. eLife, 3, e01741.CrossRefGoogle ScholarPubMed
Jönsson, H., Heisler, M. G., Shapiro, B. E., Meyerowitz, E. M., & Mjolsness, E. (2006). An auxin-driven polarized transport model for phyllotaxis. Proceedings of the National Academy of Sciences of the United States of America, 103, 16331638.Google ScholarPubMed
Keinath, N. F., Waadt, R., Brugman, R., Schroeder, J. I., Grossmann, G., Schumacher, K., & Krebs, M. (2015). Live cell imaging with R-GECO1 sheds light on flg22- and chitin-induced transient [Ca2+]cyt patterns in Arabidopsis. Molecular Plant, 8, 11881200. https://doi.org/10.1016/j.molp.2015.05.006.CrossRefGoogle ScholarPubMed
Kelner, A., Leitão, N., Chabaud, M., Charpentier, M., & de Carvalho-Niebel, F. (2018). Dual color sensors for simultaneous analysis of calcium signal dynamics in the nuclear and cytoplasmic compartments of plant cells. Frontiers in Plant Science, 9, 245.CrossRefGoogle ScholarPubMed
Korsbo, N., & Jönsson, H. (2020). It’s about time: Analysing simplifying assumptions for modelling multi-step pathways in systems biology. PLoS Computational Biology, 16, e1007982. https://doi.org/10.1371/journal.pcbi.1007982.CrossRefGoogle ScholarPubMed
Lambert, T. J. (2019). FPbase: A community-editable fluorescent protein database. Nature Methods, 16, 277278.CrossRefGoogle ScholarPubMed
Leitão, N., Dangeville, P., Carter, R., & Charpentier, M. (2019). Nuclear calcium signatures are associated with root development. Nature Communications, 10, 4865. https://doi.org/10.1038/s41467-019-12845-8.CrossRefGoogle ScholarPubMed
Liao, C. Y., Smet, W., Brunoud, G., Yoshida, S., Vernoux, T., & Weijers, D. (2015). Reporters for sensitive and quantitative measurement of auxin response. Nature Methods, 12, 207210.CrossRefGoogle ScholarPubMed
Lim, S.-L., Voon, C. P., Guan, X., Yang, Y., Gardeström, P., & Lim, B. L. (2020). In planta study of photosynthesis and photorespiration using NADPH and NADH/NAD+ fluorescent protein sensors. Nature Communications, 11, 3238.CrossRefGoogle ScholarPubMed
Long, F., Zhou, J., & Peng, H. (2012). Visualization and analysis of 3D microscopic images. PLoS Computational Biology, 8, e1002519. https://doi.org/10.1371/journal.pcbi.1002519.CrossRefGoogle ScholarPubMed
Mahlandt, E. K., & Goedhart, J. (2021). Visualizing and quantifying data from timelapse imaging experiments. BioRxiv, 2021.02.24.432684.Google Scholar
Mellor, N. L., Voß, U., Janes, G., Bennett, M. J., Wells, D. M., & Band, L. R. (2020). Auxin fluxes through plasmodesmata modify root-tip auxin distribution. Development (Cambridge, England), 147, dev181669. https://doi.org/10.1242/dev.181669.CrossRefGoogle ScholarPubMed
Miyawaki, A., Llopis, J., Heim, R., McCaffery, J. M., Adams, J. A., Ikura, M., & Tsien, R. Y. (1997). Fluorescent indicators for Ca2+ based on green fluorescent proteins and calmodulin. Nature, 388, 882887. https://doi.org/10.1038/42264.Google ScholarPubMed
Nakajima, K., Sena, G., Nawy, T., & Benfey, P. N. (2001). Intercellular movement of the putative transcription factor SHR in root patterning. Nature, 413, 307311.CrossRefGoogle ScholarPubMed
Prigge, M. J., Platre, M., Kadakia, N., Zhang, Y., Greenham, K., Szutu, W., Pandey, B. K., Bhosale, R. A., Bennett, M. J., Busch, W., & Estelle, M. (2020). Genetic analysis of the Arabidopsis TIR1/AFB auxin receptors reveals both overlapping and specialized functions. eLife, 9, e54740. https://doi.org/10.7554/eLife.54740.CrossRefGoogle ScholarPubMed
Raissig, M. T., Matos, J. L., Gil, M. X. A., Kornfeld, A., Bettadapur, A., Abrash, E., Allison, H. R., Badgley, G., Vogel, J. P., Berry, J. A., & Bergmann, D. C. (2017). Mobile MUTE specifies subsidiary cells to build physiologically improved grass stomata. Science, 355, 12151218. https://doi.org/10.1126/science.aal3254.CrossRefGoogle ScholarPubMed
Rizza, A., Tang, B., Stanley, C. E., Grossmann, G., Owen, M. R., Band, L. R., & Jones, A. M. (2021). Differential biosynthesis and cellular permeability explain longitudinal gibberellin gradients in growing roots. Proceedings of the National Academy of Sciences, 118, e1921960118. https://doi.org/10.1073/pnas.1921960118.CrossRefGoogle ScholarPubMed
Rizza, A., Walia, A., Lanquar, V., Frommer, W. B., & Jones, A. M. (2017). In vivo gibberellin gradients visualized in rapidly elongating tissues. Nature Plants, 3, 803813.CrossRefGoogle ScholarPubMed
Rizza, A., Walia, A., Tang, B., & Jones, A. M. (2019). Visualizing cellular gibberellin levels using the nlsGPS1 Förster resonance energy transfer (FRET) biosensor. Journal of Visualized Experiments, 2019, 58739.Google Scholar
Rowe, J. H, Rizza, A., Jones, A. M. (2021) Quantifying phytohormones in vivo with FRET biosensors and the FRETENATOR analysis toolset. Methods in Molecular Biology, in press.Google Scholar
Rowe, J. H., Topping, J. F., Liu, J., & Lindsey, K. (2016). Abscisic acid regulates root growth under osmotic stress conditions via an interacting hormonal network with cytokinin, ethylene and auxin. New Phytologist, 211, 225239.CrossRefGoogle ScholarPubMed
Schneider, C. A., Rasband, W. S., & Eliceiri, K. W. (2012). NIH Image to Image J: 25 years of image analysis. Nature Methods, 9, 671675. https://doi.org/10.1038/nmeth.2089.CrossRefGoogle Scholar
Schwarzländer, M., Fricker, M. D., Müller, C., Marty, L., Brach, T., Novak, J., Sweetlove, L. J., Hell, R., & Meyer, A. J. (2008). Confocal imaging of glutathione redox potential in living plant cells. Journal of Microscopy, 231, 299316. https://doi.org/10.1111/j.1365-2818.2008.02030.x.CrossRefGoogle ScholarPubMed
Stadler, R., Wright, K. M., Lauterbach, C., Amon, G., Gahrtz, M., Feuerstein, A., Oparka, K. J., & Sauer, N. (2004). Expression of GFP-fusions in Arabidopsis companion cells reveals non-specific protein trafficking into sieve elements and identifies a novel post-phloem domain in roots. The Plant Journal, 41, 319331. https://doi.org/10.1111/j.1365-313X.2004.02298.x.Google Scholar
Ulmasov, T., Murfett, J., Hagen, G., & Guilfoyle, T. J. (1997). Aux/IAA proteins repress expression of reporter genes containing natural and highly active synthetic auxin response elements. Plant Cell, 9, 19631971.Google ScholarPubMed
Vernon, I., Liu, J., Goldstein, M., Rowe, J., Topping, J., & Lindsey, K. (2018). Bayesian uncertainty analysis for complex systems biology models: Emulation, global parameter searches and evaluation of gene functions. BMC Systems Biology, 12, 1.CrossRefGoogle ScholarPubMed
Waadt, R., Hitomi, K., Nishimura, N., Hitomi, C., Adams, S. R., Getzoff, E. D., & Schroeder, J. I. (2014). FRET-based reporters for the direct visualization of abscisic acid concentration changes and distribution in Arabidopsis. eLife, 3, e01739.CrossRefGoogle ScholarPubMed
Waadt, R., Köster, P., Andrés, Z., Waadt, C., Bradamante, G., Lampou, K., Kudla, J., & Schumacher, K. (2020). Dual-reporting transcriptionally linked genetically encoded fluorescent indicators resolve the spatiotemporal coordination of cytosolic abscisic acid and second messenger dynamics in Arabidopsis. Plant Cell, 32, 25822601. https://doi.org/10.1105/tpc.19.00892.CrossRefGoogle ScholarPubMed
Waadt, R., Krebs, M., Kudla, J., & Schumacher, K. (2017). Multiparameter imaging of calcium and abscisic acid and high-resolution quantitative calcium measurements using R-GECO1-mTurquoise in Arabidopsis. New Phytologist, 216, 303320. https://doi.org/10.1111/nph.14706.CrossRefGoogle ScholarPubMed
Walia, A., Waadt, R., & Jones, A. M. (2018). Genetically encoded biosensors in plants: Pathways to discovery. Annual Review of Plant Biology, 69, 497524.CrossRefGoogle ScholarPubMed
Weigert, M., Schmidt, U., Haase, R., Sugawara, K., & Myers, G. (2020). Star-convex polyhedra for 3D object detection and segmentation in microscopy. In The IEEE Winter Conference on Applications of Computer Vision (WACV) (pp. 36553662). IEEE. https://doi.org/10.1109/WACV45572.2020.9093435.CrossRefGoogle Scholar
Wolny, A., Cerrone, L., Vijayan, A., Tofanelli, R., Barro, A. V., Louveaux, M., Wenzl, C., Strauss, S., Wilson-Sánchez, D., Lymbouridou, R., Steigleder, S. S., Pape, C., Bailoni, A., Duran-Nebreda, S., Bassel, G., Lohmann, J. U., Tsiantis, M., Hamprecht, F. A., Schneitz, K., … Kreshuk, A. (2020). Accurate and versatile 3D segmentation of plant tissues at cellular resolution. eLife, 9, 134. https://doi.org/10.7554/eLife.57613.CrossRefGoogle ScholarPubMed
Wu, R., Duan, L., Pruneda-Paz, J. L., Oh, D.-H., Pound, M., Kay, S., & Dinneny, J. R. (2018). The 6xABRE synthetic promoter enables the spatiotemporal analysis of ABA-mediated transcriptional regulation. Plant Physiology, 177, 16501665.CrossRefGoogle ScholarPubMed
Zhang, L., Takahashi, Y., Hsu, P. K., Kollist, H., Merilo, E., Krysan, P. J., & Schroeder, J. I. (2020). FRET kinase sensor development reveals SnRK2/OST1 activation by ABA but not by MeJA and high CO2 during stomatal closure. eLife, 9, 174. https://doi.org/10.7554/eLife.56351.CrossRefGoogle Scholar
Zürcher, E., Tavor-Deslex, D., Lituiev, D., Enkerli, K., Tarr, P. T., & Müller, B. (2013). A robust and sensitive synthetic sensor to monitor the transcriptional output of the cytokinin signaling network in planta. Plant Physiology, 161, 10661075.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Different types of genetically encoded fluorescent biosensors and their molecular mechanisms. (a) The mechanism of action of the auxin reporter DR5::GFP, where the auxin receptor complex targets the AUX/IAA transcriptional repressors for degradation, allowing green fluorescent protein transcription and fluorescence in the presence of the hormone. (b) The mechanism of action of the auxin biosensor DII:VENUS, where a venus fluorescent protein (FP) fused to domain II (DII) from an AUX/IAA protein is targeted for degradation by the auxin receptor complex, causing loss of fluorescence in the presence of the hormone. (c) The mechanism of action of the direct abscisic acid biosensor ABACUS1, where hormone binding causes the interaction of two sensory domains of a single fusion protein. This conformational change causes increased Förster resonance energy transfer (FRET) between two FP domains (a FRET donor and acceptor) and an altered emission ratio upon donor excitation. (d) The mechanism of action of the direct glutathione redox potential biosensor roGFP. A disulphide bond on the barrel of roGFP is reversibly sensitive to oxidation, causing an altered absorption spectrum.

Figure 1

Fig. 2. Example image processing pipeline for the nuclear localised ratiometric Förster resonance energy transfer sensor nlsABACUS1-2μ. Notes: Several image acquisition channels are used in FRET biosensor analysis. Here, the new FRETENATOR analysis pipeline for Fiji was used (Rowe et al., 2021). For segmentation, the following steps are performed: first a difference of Gaussian filter is applied to the acceptor-excited acceptor-emission channel to remove background and smooth noise, then Otsu’s method is used to threshold the filtered image based on signal intensity. A watershed algorithm is used to split touching objects in the threshold image, and a connected-component analysis is used to label all the individual objects, producing a label map. The quantification steps involve removing saturated voxels from the original images and then dividing the mean signal intensity of donor-excited acceptor-emission channel by the donor-exited donor-emission channel for each labelled ROI, which can be represented on the segmented image using false coloration (e.g., the Turbo lookup table used here).

Author comment: Focus on biosensors: Looking through the lens of quantitative biology — R0/PR1

Comments

No accompanying comment.

Review: Focus on biosensors: Looking through the lens of quantitative biology — R0/PR2

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: The review by Rowe and Jones provides an overview of genetically encoded fluorescent biosensors (GEFBs) and recent advances in the field. The authors also provide examples of quantitative analysis using GEFBs and discuss well the problems of quantitative research. However, the reviewer unfortunately could not figure out from the text what the “two of the main bottlenecks” the authors mentioned in the abstract were. Also, the word “this” is often used throughout the text, but it is often unclear what it is referring to. Please revisit it. The figure is too small to see the text, and it is hard to understand.

There are also several minor points and editorial comments that may help to improve the clarity of this review further.

Page 2.

“DII-Venus ubiquitination causes rapid degradation, reducing the time delay between hormone accumulation and fluorescence response.”

The author later stated that these sensors cannot be used to image fast hormone depletion or oscillations, so it would be helpful if the author could provide the time required for these reactions.

“A powerful tool to overcome this problem is the expression of a second fluorescent protein, as in the R2D2 or qDII reporters (Figure 1B).”

These reporters are not described in Figure 1B.

“Many also have a ratiometric output to control for optical artefacts during imaging and differences in expression.”

I did not follow what optical artefacts mean. It would be helpful to the reader if the authors provide examples with appropriate papers.

Page 2. “Herud-Sikimic et al., 2020” bioRxiv paper was published in Nature 2021. The reference should be changed.

Page 3. “Band, Úbeda-Tomás et al., 2012” does not show the multiscale modeling based on the expression of the GA20ox biosynthesis genes. Please refer to the appropriate paper or rephrase the sentence.

“Because this contrasted with the pattern observed with the nlsGPS1 FRET biosensor,”

It is unclear what “the pattern” means. It would be clearer what the pattern is if the author explains what the nlsGPS1 FRET biosensor is. It would also make the text easier to understand if the author could be more specific about what “this” refers to.

“The model predicted that increasing permeability along the root would eliminate the gradient when exogenous hormone is applied, which could be tested in vivo by applying gibberellin at low exogenous pH and indeed resulted in a loss of the gradient.”

It would be helpful to the reader if the author could describe this in more detail. Why does the low exogenous pH increase the permeability of gibberellins along the root? Doesn’t the low pH affect the sensor itself?

Page 4.

“Recent pioneering co-expression of pair of direct biosensors has allowed the temporal dynamics of many second messenger or hormones to be characterized simultaneously (Waadt et al., 2017, 2020). This has confirmed many predictions and offered new insight.”

It would be better to write specifically about which second messenger and hormone dynamics are actually observable in the paper instead of using “many”, which predictions have been clarified by these technologies, and what new insights it has brought.

Minor edits:

Page 2.

mean → means

hormone depletions → hormone depletion

Page 3.

difficulty measuring → difficulty of measuring

real time → real-time

modelling determine → modelling to determine

Recommendation: Focus on biosensors: Looking through the lens of quantitative biology — R0/PR3

Comments

Comments to Author: Dear Authors

Thank you for submitting to QPB. We received the reviewers from two external reviewers, of whom one reviewer prefer not to reveal his/her identity. Both reviewers gave valuable and constructive advice on how to improve the quality of the manuscript further.

Below are the comments of the anonymous reviewer:

The manuscript entitled „Focus on biosensors: looking through the lens of quantitative biology” by Rowe and Jones reviews, summarizes, highlights, some characteristics, applications, new aspects and future directions of genetically encoded biosensor research in plants. The manuscript is well written and apart from some small inaccuracies (mentioned below), I enjoyed reading it. I am missing a bit the concept of this article, because sometimes I had the feeling that the authors just throwed in some bullet points. The context and connections of the different sections was not clearly visible. Also, the discrimination between indirect transcriptional reporters and direct sensors was not precise. The terms extrinsic and intrinsic sensors and the explanation of this came too late. Maybe explain first the difference between indirect and direct and intrinsic and extrinsic biosensors, before discussing them in further detail.

Minor comments:

1) For helping the reviewer to properly comment on the article and the author to find the respective text, it is strongly recommended to also include line numbers to the manuscript.

2) “In recent years, plant biologists have started to transition from transcriptional reporter systems to genetically encoded fluorescent biosensors (GEFBs).” Transcriptional reporter systems, promoter:GFP reporters, also fall into the category of GEFBs. They are also genetically encoded. Please specify better or remove “transition from”.

3) “Biosensors allow the in vivo quantification of biochemistry…” Not all biosensors allow this. Please be more precise. Maybe use GEFBs here.

4) Although the abstract is clearly written, the text just provides some bullet points about Biosensors and GEFBs. The scope of this article is however not clear.

5) “These are proteins that contain one or more FPs, …”. The definition of GEFBs is a bit vague. Please be more precise.

6) “… see our recent review…” Remove “our”, as the first author of this article is not a co-author of this citation. Better stay neutral with such statements.

7) “From transcriptional reporters to genetically encoded biosensors”. Transcriptional reporters also belong to genetically encoded fluorescent biosensors. Maybe discriminate by direct and in-direct biosensors. Transcriptional reporters are indirect, FRET sensors are direct etc…

8) “… but at best, transcriptional reporters such as DR5 indicate where hormones were.”. These reporters do not detect the hormone itself, but the hormone response, so they indicate where the hormone response is strongest compared to other tissues.

9) “… due to changes in expression and degradation.”: Of what?

10) “By comparing the DII-nVenus fluorescence”. Please specify what the “n” stands for. Do you mean “m” for monomeric?

11) “…proteasomal machinery elicit fluorescence…” elicited?

12) “However, DII:Venus, R2D2 and qDII…” This sentence appears incomplete.

13) “… these are the direct GEFBs”. Good, so also define the indirect GEFBs above.

14) “…the first plant hormone GEFB were…” GEFBs?

15) “Herus-Sikimic et al., 2020” This is a not yet peer-reviewed article. Also, by looking at the properties of this GEFBs and the content of the manuscript, in my opinion this article is not yet worth to be cited.

16) “… from the donor CFP…” Better use here “CFP variant”, as CFP is not used in ABACUS1-2µ.

17) “Hanson et al., 2004”. Maybe include also a reference for the first use of roGFP in plants.

18) “nlsGPS1 FRET biosensor” Good to also state here that this sensor is GA responsive/sensitive.

19) mathematical modeling “to” determine…

20) “Rather than the presumes…” Make clear that you are speaking in this paragraph about GA in Arabidopsis roots.

21) “a” behaviour that could only be…

22) “…increasing permeability…” of what?

23) “… the gradient…” of what?

24) “…many biological processes involved…” “involve”?

25) “Huang et al., 2019” is relevant for guard cells, but used the biosensor developed by Waadt et al., 2017. In this context maybe worth to cite also Keinath et al., 2015.

26) “Leitao et al., 2019”. In this context maybe cite also Kelner er at., 2018. Leitao used the biosensors that were developed by Kelner.

27) “Exposito-Rodriguez et al., 2017”. Not sure about this reference. Experiments were done after transient expression in tobacco cells with sensors that not really work in transgentic Arabidopsis plants. For subcellular ROS/redox, better cite work from Markus Schwarzländer.

28) Regarding image analysis and data processing: Another bottleneck forgotten to be mentioned by the authors is focal drifts, or ROI drifts due to continuous plant growth during biosensor imaging. How to compensate for this?

29) At some point in the manuscript maybe include a small paragraph about light-sheet microscope-based biosensor imaging.

Figure 1) Add a title. If not cited in the main text, cite also the other biosensors mentioned in the figure. ABACUS topology is wrong (in c), see citation from original article Fig 3a:” The overall domain order of ABACUS1 is N-terminus–edCitrine–attB1–ABI1aid–L52–PYL1–attB2–edCerulean–C-terminus.”. If you don’t want to change the topology, you should indicate that the N-terminus is at the acceptor site and the C-terminus at the donor site.

Please revise your manuscript according to the advice of both reviewers.

Yours sincerely

Boon Leong LIM

Decision: Focus on biosensors: Looking through the lens of quantitative biology — R0/PR4

Comments

No accompanying comment.

Author comment: Focus on biosensors: Looking through the lens of quantitative biology — R1/PR5

Comments

We are pleased to submit our revised manuscript for consideration after addressing the very helpful comments of the editor and reviewer.

Review: Focus on biosensors: Looking through the lens of quantitative biology — R1/PR6

Comments

Comments to Author: I appreciate the responses and revised manuscript. The changes made by the author led to an improvement of this review.

Minor comments:

"Band, Úbeda-Tomás, et al., 2012" is not listed in the reference list.

Line 212: “difficulty measuring” difficulty of measuring?

Line 271: “recently was recently” Either recently should be removed.

Line 276: “This is perhaps” It may be unclear what This refers to.

Recommendation: Focus on biosensors: Looking through the lens of quantitative biology — R1/PR7

Comments

Comments to Author: Dear Authors

Thank you for submitting to QPB. We received the reviewers from two external reviewers, of whom one reviewer prefer not to reveal his/her identity. Both reviewers suggested minor revision before acceptance.

Below are the comments of the anonymous reviewer:

The revised manuscript by Rowe and Jones entitled “Focus on biosensors: looking through the lens of quantitative biology”, has been significantly improved. The concept of analyzing GEFBs using state-of-the-art image analysis pipelines and feeding the data into computational models is recognizable and the manuscript is overall much better sorted.

There are a few minor things that came to my attention and that could be improved:

Line 48, 83, 123, 137 and several more) sort publications by publication date (oldest first) and alphabetically.

Line 51) This jump from “stress responses” to “FP maturation time” is very disturbing and somehow leads to a very abrupt topic change of this section. I recommend revising this section so that it has a clear focus. Biological findings made with transcriptional reporters, or low temporal resolution of transcriptional reporters due to slow maturation times and long half-lives of FPs.

Line 204) such as?

Line 271) “recently” doubled.

Line 302) where a Venus FP?

Line 320) threshold the filtered image

Reviewer 1

I appreciate the responses and revised manuscript. The changes made by the author led to an improvement of this review.

Minor comments:

"Band, Úbeda-Tomás, et al., 2012" is not listed in the reference list.

Line 212: “difficulty measuring” difficulty of measuring?

Line 271: “recently was recently” Either recently should be removed.

Line 276: “This is perhaps” It may be unclear what This refers to.

Decision: Focus on biosensors: Looking through the lens of quantitative biology — R1/PR8

Comments

No accompanying comment.

Author comment: Focus on biosensors: Looking through the lens of quantitative biology — R2/PR9

Comments

We’re pleased to submit a final revision following acceptance for this invited commentary and thank the reviewers and editors for their helpful comments.

Recommendation: Focus on biosensors: Looking through the lens of quantitative biology — R2/PR10

Comments

Comments to Author: Dear Dr. Jones

We are pleased to inform you that your article "Focus on biosensors: looking through the lens of quantitative biology" has been accepted for publication in Quantitative Plant Biology.

Thank you for choosing QPB to publish your work, we look forward to receiving further contributions from your research group in the future.

Yours sincerely

Boon Leong LIM

Associate Editor

Decision: Focus on biosensors: Looking through the lens of quantitative biology — R2/PR11

Comments

No accompanying comment.