We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
ReportsOpen Accesscc iconby iconnc iconnd icon

A simplified and rapid in situ hybridization protocol for planarians

    &
    Ryan S King

    *Author for correspondence: Tel.: +1 920 403 3237;

    E-mail Address: ryan.king@snc.edu

    St. Norbert College, De Pere, WI 54115, USA

    Published Online:https://doi.org/10.2144/btn-2023-0074

    Abstract

    Whole-mount in situ hybridization is a critical technique for analyzing gene expression in planarians. While robust in situ protocols have been developed, these protocols are laborious, making them challenging to incorporate in an academic setting, reducing throughput and increasing time to results. Here, the authors systematically tested modifications to all phases of the protocol with the goal of eliminating steps and reducing time without impacting quality. This modified protocol allows for whole-mount colorimetric in situ hybridization and multicolor fluorescence in situ hybridization to be completed in two days with a significant reduction in steps and hands-on processing time.

    METHOD SUMMARY

    A planarian in situ hybridization protocol was optimized for time and simplicity. Modifications to the fixation, bleaching and blocking solutions were tested to identify conditions that reduced time and number of steps without impacting staining quality.

    Gene expression analysis is a foundational tool in cell and developmental biology studies. Next-generation sequencing technologies have revolutionized the breadth and resolution for examining the molecular profile of cells and how they change during development or in response to stimuli. However, validating and following up on interesting leads from sequencing projects requires low-throughput techniques. In situ hybridization (ISH) is an established method for analyzing expression patterns that provides valuable spatial information for understanding the context of gene expression. Protocols for ISH are laborious as they must accomplish several important goals. First, fixation: mRNA molecules must be locked in position and tissue morphology preserved. Second, permeabilization: staining reagents need access to target molecules. Third, hybridization: chemically modified RNA probes need to specifically hybridize to target mRNA molecules. Finally, detection: methods for visualizing the location of probe binding must provide a strong signal with minimal background staining. Numerous protocols have been developed for ISH in diverse organisms [1–4] and while there are often species-specific techniques, many of the processes and reagents are shared across model systems.

    Planarians have emerged as a valuable model system for studying stem cell biology, regeneration and other foundational topics in biology [5]. Planarians present some unique challenges for ISH and immunostaining techniques: they secrete a layer of mucus that must be removed prior to fixation to allow penetration of staining reagents and many species produce body pigment that can hinder visualization in staining experiments. Previous work has focused on developing reliable protocols with improved spatial resolution and sensitivity of staining [1,6–8]. Like protocols for other model organisms, ISH protocols for planarians are laborious, multiday procedures that limit throughput and are challenging to incorporate into laboratory experiences for undergraduate students. Here, the authors focused on modifying, combining or eliminating steps of the procedure to reduce the overall time and number of contact points with samples.

    Materials & methods

    Animal husbandry

    Asexual Schmidtea mediterranea clonal line CIW4 and Dugesia japonica were maintained as previously described [7].

    Probe synthesis

    Genes of interest were PCR-amplified from cDNA and cloned into pJC53.2 as previously described [9]. The DNA template for probe synthesis was generated by PCR amplification using T7 primers and purified using a DNA clean and concentrator kit (Zymo Research, CA, USA). Antisense RNA probes were synthesized by in vitro transcription with either DIG-11-UTP (Sigma-Aldrich, MO, USA) or Fluorescein-12-UTP (Sigma-Aldrich) using either the T3 or SP6 riboprobe system (Promega, WI, USA) with modifications from the manufacturer's suggested protocol. The transcription reaction volume was reduced to 10 μl and 0.5 units of thermostable inorganic pyrophosphatase (New England Biolabs, MA, USA) was included in the reaction. Following the transcription reaction, the DNA template was degraded by treatment with DNase (Promega, WI, USA) before being analyzed on a 1% agarose gel. Successful reactions were diluted 1:100 in prehybridization solution and stored at -20°C.

    In situ hybridization

    The protocol from King and Newmark, 2013 [7], served as a starting reference against which to compare protocol alterations. Modifications to the protocol included the addition of 20% methanol, 10% acetic acid and 5 mM ethylenediaminetetraacetic acid to the fixative; increasing the H2O2 concentration to 6% and adding detergent to the bleach solution; elimination of the postfixation dehydration and proteinase K steps; reduced washing times and steps; inclusion of cold-water fish gelatin in the blocking solution; room temperature antibody incubation; elimination of polyvinyl alcohol (PVA) from the development solution; and colorimetric development at 37°C.

    Immunofluorescence & FISH

    Samples were processed using the rapid protocol with probes visualized using tyramide signal amplification as previously described [7]. M-phase cells were labeled using 1:5000 antiphospho-histone H3 (Cell Signaling Technology, MA, USA) and 1:1000 antirabbit-HRP (Jackson ImmunoResearch Laboratories, PA, USA) and detected by tyramide signal amplification. Ciliated protonephridia were labeled with 1:1000 antiacetylated-alpha tubulin (Santa Cruz Biotech, TX, USA) and 1:500 Alexa 488 antimouse (Thermo Fisher Scientific, MA, USA). Muscle was labeled with 1:50 6G10 antibody (Developmental Studies Hybridoma Bank, IA, USA) and 1:1000 antimouse-HRP (Jackson ImmunoResearch Laboratories) followed by tyramide signal amplification. For BrdU labeling, animals were soaked in 20 mg/ml 5-bromo-2′-deoxyuridine with 3% dimethyl sulfoxide in a 6 g/l Instant Ocean salt solution for 2 h followed by a 4-h chase prior to processing using the rapid protocol. BrdU incorporation was detected using 1:25 anti-BrdU antibody (BD Biosciences, NJ, USA) and 1:1000 antimouse-HRP (Jackson ImmunoResearch Laboratories, PA, USA) followed by tyramide signal amplification.

    Multicolor FISH

    Samples were hybridized with digoxigenin and fluorescein-labeled probes for the genes of interest. Sequential detection using two rounds of tyramide signal amplification was performed as previously described [7] with a 100 mM azide quenching step between rounds of amplification. Simultaneous detection was performed by incubating with both 1:2000 antidig-AP (Sigma-Aldrich) and 1:2000 antifluorescein-POD (Sigma-Aldrich), followed by tyramide signal amplification and development using the fluorescent alkaline phosphatase substrate Fast Blue BB (Sigma-Aldrich) as described [8,10].

    Imaging

    Samples were cleared in 80% glycerol and mounted on slides. Images of colorimetric samples were collected with an Olympus SZ61TR stereomicroscope (Olympus, Tokyo, Japan) with an AmScope MU1803-HS camera using AmLite software (AmScope, CA, USA). Fluorescent samples were imaged using an Olympus Fluoview FV1200 laser scanning confocal microscope (Olympus) and images were processed using FIJI software [11].

    Results & discussion

    Modification of fixation conditions eliminates enzymatic permeabilization steps for high-abundance transcripts

    The sensitivity and reliability of ISH in planarians have been the focus of several studies [1,6–8]. While published protocols produce samples with good morphology and strong signal-to-background (Figure 1A & B) [7], they are lengthy, with numerous sample processing steps, and require a few days to complete. We wondered whether ISH could be optimized for time with minimal reduction in sample quality and signal strength. Two key parameters that need to be balanced for successful ISH are fixation and permeabilization. mRNA molecules must be retained in position and morphology maintained while staining reagents need to penetrate deep tissues of the sample for labeling. Fixation of planarians with 4% formaldehyde without further postfixation dehydration and enzymatic permeabilization steps resulted in a weak signal with poor staining in the prepharyngeal region (Figure 1C & D). As was shown for other model systems [12], the addition of acetic acid to the fixative modulates formaldehyde crosslinking, eliminating the need for additional permeabilization steps (Figure 1E & F). The incorporation of acetic acid in the fixative likely contributes more than simply attenuating crosslinking, as reducing formaldehyde concentration or quenching crosslinking using a Tris buffer [13] increases sample fragility with mixed effects on signal intensity (Supplementary Figure 1). The inclusion of methanol in the fixative further improves the consistency and intensity of staining (Figure 1G & H). The modified fixative also seems to increase the flexibility in timing for mucus removal using the mucolytic N-acetyl-l-cysteine (NAC) with only modest improvements for longer NAC incubation times (Supplementary Figure 2).

    Figure 1. Addition of acetic acid and methanol to fixative eliminates requirement for postfixation methanol dehydration and proteinase K permeabilization.

    (A & B)In situ hybridization for (A)smedwi-1 and (B)Smed-porcn-1 following fixation in 4% formaldehyde with postfixation methanol dehydration and proteinase K permeabilization steps. (C & D) Signal is greatly reduced following fixation in 4% formaldehyde solution with postfix methanol dehydration but absence of proteinase K treatment. (E & F) Incorporation of acetic acid into fixative increases signal in absence of methanol dehydration and proteinase K treatment. (G & H) Increasing methanol concentration in fixative to 20% slightly increases signal and provides more reliable staining in prepharyngeal region in absence of a separate methanol dehydration and proteinase K treatment. Scale bar = 500 μm.

     Similar levels of in situ signal and background with high-abundance transcripts were found for both the rapid in situ protocol and samples fixed with 4% formaldehyde and permeabilized using proteinase K as previously described (Supplementary Figure 3A & D). However, for several low-abundance transcripts, the in situ signal was weak or absent using the rapid protocol (Supplementary Figure 3B & C). There could be several factors leading to the difference in sensitivity of the rapid protocol. The fixation conditions for the rapid protocol may not adequately fix the transcripts in place, allowing for their diffusion or degradation during processing. Alternatively, the rapid protocol may not sufficiently permeabilize the tissue, allowing adequate access to the transcripts. Signal sensitivity may vary for transcripts using the rapid protocol depending on their abundance, cell type of expression or RNA structure, such as inclusion in ribonucleoprotein granules. To increase the utility of the rapid protocol, we examined whether combining rapid bleaching and posthybridization processing with modified fixation and permeabilization steps could improve sensitivity while maintaining a significant reduction in protocol time. We found greatly improved sensitivity by performing a 7.5-min NAC treatment, fixing in 4% formaldehyde solution, dehydrating in 100% methanol, bleaching for 1 h in 6% H2O2 (no detergent) and performing a 10-min proteinase K permeabilization step prior to hybridization and processing using the rapid protocol (Supplementary Figure 3E & F). We also examined whether adding a proteinase K permeabilization step to the rapid protocol after bleaching improved sensitivity. To prevent samples from disintegrating during the proteinase K step, we performed the bleaching step in 6% H2O2 without detergent and did not agitate the samples during the proteinase K incubation. While the inclusion of the proteinase K step in the rapid protocol improved sensitivity for low-abundance transcripts compared with the rapid protocol alone, sensitivity was sometimes reduced compared with fixation in 4% formaldehyde (Supplementary Figure 3E & H), and sometimes improved (Supplementary Figure 3F & I). Based on the genes we tested, 4% formaldehyde fixation and proteinase K permeabilization combined with the hybridization and wash steps of the rapid protocol is a suitable modification for genes where the rapid protocol alone is not sufficiently sensitive.

    Increasing bleaching strength improves permeability

    Several hydrogen peroxide-based bleaching solutions have been described for bleaching body pigment in planarians [1,6,7], including a 1.2% H2O2/formamide bleach capable of producing greater sensitivity of staining for low-abundance genes [7]. However, this formamide-based bleaching solution requires a couple of hours of incubation to achieve full bleaching and, at shorter incubation times, not only does pigmentation remain, but the staining intensity is diminished in the absence of an enzymatic permeabilization step (Figure 2A–C). Increasing the concentration of H2O2 in the formamide solution to 6% reduced bleaching time and improved signal but, occasionally, samples showed weak staining in the prepharyngeal region (Figure 2D–F). Enzymatic permeabilization steps not only use proteinase K to improve staining but also detergents such as sodium dodecyl sulfate (SDS). Adding SDS and Triton X-100 to the 6% H2O2 formamide bleaching solution improved the consistency of staining in the prepharyngeal region (Figure 2G–I).

    Figure 2. Increasing hydrogen peroxide concentration and addition of detergents in bleaching solution improves permeability.

    (A–C) Planarians bleached in a formamide bleaching solution containing 1.2% hydrogen peroxide show residual pigmentation after 1 h of bleaching and reduced signal for (A)Smed-porcn-1(B), smedwi-1 and (C)Smed-CAVII-1. (D–F) Bleaching in a 6% hydrogen peroxide formamide bleaching solution eliminates pigmentation after 1 h and improves signal. (G–I) Addition of sodium dodecyl sulfate and triton X-100 to 6% hydrogen peroxide formamide bleaching solution slightly improves staining in prepharyngeal region. Scale bar = 500 μm.

    Optimizing blocking & wash steps

    Solution changes and wash steps are one of the most laborious aspects of ISH with each processing step increasing the chances of damaging samples. Therefore, we set out to test if extensive wash steps are necessary for reducing background staining. Effective washing requires a difference in the concentration of components between the sample and the wash solution and time for diffusion of components in or out of the sample. We reasoned that equilibrium between the sample and wash solution occurs quickly for the first change to a new solution as the residual liquid left behind in the tube gets rapidly diluted in the newly added wash solution and that a few short washes followed by longer washes might be sufficient for reducing background. We tested several washing methods to identify the most efficient for reducing background staining (Figure 3). Mock hybridized planarians incubated with blocking solution lacking anti-DIG-AP antibody showed no background staining after 4 h of development (Figure 3A & B). Samples washed one time for <1 min prior to development showed a significant amount of background staining (Figure 3C & D). Samples washed with 4 solution changes for a total of 35 min showed only slightly more background staining compared with the recommended 9 solution changes over 2.25 h (Figure 3E–H) [7], indicating that wash time and steps could be substantially reduced.

    Figure 3. Incorporation of fish gelatin into blocking solution may reduce background while decreasing wash time.

    (A–H) Samples incubated in hybridization buffer lacking labeled probe. (A & B) No antibody control showing absence of background staining following development. (C & D) Background staining obtained following removal of antibody solution with a <1 min wash in AP buffer prior to development. Background staining is reduced in blocking solution containing (D) 0.45% cold water fish gelatin compared with (C) blocking solution with horse serum and Roche Western Blocking Solution alone. (E–H) Number of wash steps and time can be reduced from >2 h (G & H) to <40 min (E & F) without significant background development with (F) incorporation of fish gelatin in blocking solution. (I & J) Fish gelatin does not negatively impact smedwi-1 staining. Scale bar = 500 μm.

    Even the long washing procedure still produced background compared with the no-antibody controls, suggesting that the antibody may be nonspecifically binding. Cold-water fish gelatin has been used as a blocking reagent for immunostaining experiments in planarians [14]. We tested whether the inclusion of fish gelatin in the blocking solution could reduce background staining and found a significant reduction in background compared with the blocking solution containing horse serum and Roche Western Blotting Reagent alone (Figure 3A–H). The addition of fish gelatin to the blocking solution did not inhibit the binding of the antibody to its target, as development time and signal intensity were similar whether it was present in the blocking solution or not (Figure 3I & J). Antifluorescein-POD antibodies used for FISH show significant background staining that can be reduced by the inclusion of a Roche Western Blocking Reagent in the blocking and antibody solution [7]. We examined whether also including fish gelatin in the blocking and antibody solution would further improve background staining, but found comparable levels of background with and without fish gelatin present (Supplementary Figure 4). We also did not note significant differences in background staining with strong probes between blocking solutions with and without fish gelatin, but the results indicate the inclusion of fish gelatin may be particularly beneficial for the detection of low-abundance genes.

    With the maintained quality and time savings found by reducing postantibody wash steps, we tried reducing other wash steps, particularly posthybridization. We reasoned that any unhybridized probe remaining in the samples would be degraded by nucleases in the blocking solution at later steps and found that shortening these washes still generated satisfactory results (most figures show samples with reduced posthybridization washes). Additionally, quality staining can be obtained with reduced prehybridization and preantibody blocking times as well as 2-h room temperature antibody incubation times (most figures show samples with shortened blocking and antibody incubation times). These modifications provide substantial time savings, eliminating a full day off protocol time and reducing hands-on sample processing time.

    Incubation at 37°C increases rate of development

    Colorimetric development using the alkaline phosphatase substrate NBT/BCIP can take a few hours to complete. The rate of development has been shown to increase with the addition of macromolecular crowding agents such as PVA [1]. However, when testing several PVA concentrations, a significant difference in development rate between reactions containing PVA or not was not observed (Supplementary Figure 5). The viscosity of a development solution containing PVA increases the challenge of removing the solution without pipetting up or damaging samples. Furthermore, failure to remove all PVA prior to ethanol washing causes any remaining PVA to precipitate. Given the minimal benefits and additional challenges, we found it more efficient to exclude PVA from the development solution. Next, we examined whether increasing the temperature of development would reduce development time without increasing background staining. Samples incubated at 37°C showed significantly faster development, reaching an ideal staining intensity in a shorter time with no difference in signal-to-background staining (Figure 4).

    Figure 4. Incubation at 37°C increases rate of development.

    Planarians hybridized with (A–F)Smed-porcn-1 probe or (G–L)smedwi-1 probe were split after last postantibody wash and developed at room temperature (21°C) or 37°C. (A & B; G & H) Samples were collected and development stopped at 30 min, (C & D; I & J) 1 h and (E & F; K & J) 2 h. Scale bar = 500 μm.

    Rapid protocol has broad compatibility with common stains

    We next tested the versatility of the rapid ISH protocol for other common analyses. Double FISH is typically a 4-day protocol and can require substantial optimization to achieve consistent results [7]. Using the rapid protocol modifications, we were able to reduce protocol time to 3 days while maintaining strong labeling with minor background staining (Figure 5A). Immunofluorescence staining can be challenging, as fixation and processing conditions can greatly impact epitope accessibility [15,16]. We tested four commonly used antibodies for S. mediterranea (the M-phase marker phospho-histone H3 [17], the muscle marker 6G10 [16], the cilia marker acetylated-α-tubulin [18] and an antibody for detection of incorporated BrdU [14]) and found that all showed normal staining in combination with FISH using the rapid protocol for fixation and processing (Figure 5B–F). In addition, we examined whether the rapid protocol also worked for D. japonica. With slightly reduced NAC treatment concentration and time, D. japonica samples remained intact through the protocol and showed strong labeling compared with samples where mucus removal was accomplished using a 2% HCl treatment [19] (Supplementary Figure 6).

    Figure 5. Rapid protocol has broad compatibility.

    (A) Sequential double FISH using two rounds of tyramide signal amplification can be completed in three days. (B–E) Rapid FISH can be coupled with immunofluorescence staining for (B) phospho-histone H3 (Ser10), (C & D) muscle-specific antibody 6G10 and (E) acetylated alpha-tubulin. (F) Detection of BrdU in stem cells following a 4-h chase. Scale bar = 50 μm.

    To further reduce protocol time, we examined the feasibility of the simultaneous detection of two genes through a combination of tyramide signal amplification and a fluorescent alkaline phosphatase substrate, Fast Blue BB [8,10]. Using this approach, we were able to incubate with both antidig-AP and antifluorescein-POD antibodies simultaneously, allowing us to reduce the protocol time to two days (Figure 6).

    Figure 6. Two-day double FISH.

    Probe specific to proximal tubules of protonephridia was detected using fluorescein tyramide signal amplification coupled with detection of distal tubule marker Smed-CAVII-1 using fluorescent alkaline phosphatase substrate, Fast Blue BB. Scale bar = 50 μm.

    Conclusion

    ISH protocols can be substantially shortened with minimal impact on staining quality through modification of components and optimization of procedures. Permeabilization can be incorporated into fixation and bleaching steps through the addition of acetic acid and methanol to the fixative and the inclusion of detergents in the bleaching solution, thereby eliminating the need for a separate enzymatic permeabilization step. Numerous long washes are unnecessary for all but possibly the very largest samples, and wash time and labor can be saved by optimizing wash conditions and by including fish gelatin in the blocking solution. Colorimetric development time can be reduced by incubating samples at higher temperatures. In combination, these enhancements eliminate 20 sample handling steps and a full day of protocol time compared with previous methods (Table 1) [7].

    Table 1. Time and labor savings of rapid protocol compared with traditional protocol.
      Traditional protocolRapid protocol
    Day 1Time:∼423 min∼104 min
     Steps:2211
    Day 2Time:∼240 min∼270 min + 15–120 min development
     Steps:1216
    Day 3Time:∼165 min + 30–240 min development 
     Steps:13 

    Future perspective

    ISH has been a crucial tool in planarian research for analyzing gene expression. This study provides researchers with a simplified and time-efficient method for ISH using planarians. Not only will this protocol help researchers to process more samples with less labor and a quicker time to results, but it will also increase the feasibility of introducing this method in academic teaching laboratories that typically do not have schedules conducive to carrying out the traditionally lengthy protocols. Additionally, while planarians provide some unique challenges for ISH, the reagents and procedures used for planarians are generally shared with other systems and it is likely that the modifications presented here may be applicable to other model organisms.

    Executive summary
    • A time- and labor-optimized protocol for in situ hybridization in planarians was developed.

    Results & discussion

    • The inclusion of acetic acid and methanol in the fixative improves signal in the absence of enzymatic permeabilization steps.

    • Increasing hydrogen peroxide concentration and the addition of detergents to the bleaching solution reduces bleaching time and increases tissue permeability.

    • The inclusion of fish gelatin in the blocking solution reduces nonspecific antibody binding.

    • Blocking and wash steps/time can be reduced without impacting staining quality.

    • Colorimetric development at 37°C increases the rate of development.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/suppl/10.2144/btn-2023-0074

    Author contributions

    A Gaetano and R King conceived of, designed and carried out the experiments. Both authors contributed to the writing and editing of the manuscript.

    Acknowledgments

    The authors thank Kayla Koenig and Rachel McCoy for critical comments on the manuscript.

    Financial disclosure

    This work was funded by a St. Norbert College Collaborative Research Grant to A Gaetano and St. Norbert College Faculty Development and Natural Sciences divisional support to R King. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    The authors have no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Pearson BJ, Eisenhoffer GT, Gurley KA, Rink JC, Miller DE, Sánchez Alvarado A. Formaldehyde-based whole-mount in situ hybridization method for planarians. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 238(2), 443–450 (2009). • Pioneered the use of N-acetyl-L-cysteine for mucus removal and formaldehyde fixation in planarians.
    • 2. Thisse C, Thisse B. High-resolution in situ hybridization to whole-mount zebrafish embryos. Nat. Protoc. 3(1), 59–69 (2008).
    • 3. Piette D, Hendrickx M, Willems E, Kemp CR, Leyns L. An optimized procedure for whole-mount in situ hybridization on mouse embryos and embryoid bodies. Nat. Protoc. 3(7), 1194–1201 (2008).
    • 4. Legendre F, Cody N, Iampietro C et al. Whole mount RNA fluorescent in situ hybridization of Drosophila embryos. J. Vis. Exp. JoVE 71, e50057 (2013).
    • 5. Reddien PW. The cellular and molecular basis for planarian regeneration. Cell 175(2), 327–345 (2018).
    • 6. Guerrero-Hernández C, Doddihal V, Mann FG, Alvarado AS. A powerful and versatile new fixation protocol for immunohistology and in situ hybridization that preserves delicate tissues in. www.biorxiv.org/content/10.1101/2021.11.01.466817v1
    • 7. King RS, Newmark PA. In situ hybridization protocol for enhanced detection of gene expression in the planarian Schmidtea mediterranea. BMC Dev. Biol. 13(1), 8 (2013). • Provides modifications for improving the sensitivity of FISH in planarians.
    • 8. Currie KW, Brown DDR, Zhu S et al. HOX gene complement and expression in the planarian Schmidtea mediterranea. EvoDevo 7(1), 7 (2016).
    • 9. Collins JJ, Hou X, Romanova EV et al. Genome-wide analyses reveal a role for peptide hormones in planarian germline development. PLoS Biol. 8(10), e1000509 (2010).
    • 10. Lauter G, Söll I, Hauptmann G. Two-color fluorescent in situ hybridization in the embryonic zebrafish brain using differential detection systems. BMC Dev. Biol. 11(1), 43 (2011). • Describes the combination of Fast Blue BB and tyramide signal amplification for two-color FISH.
    • 11. Schindelin J, Arganda-Carreras I, Frise E et al. FIJI: an open-source platform for biological-image analysis. Nat. Methods 9(7), 676–682 (2012).
    • 12. Fernández J, Fuentes R. Fixation/permeabilization: new alternative procedure for immunofluorescence and mRNA in situ hybridization of vertebrate and invertebrate embryos. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 242(5), 503–517 (2013). • Describes the utility of incorporating acetic acid in formaldehyde fixative.
    • 13. Hoffman EA, Frey BL, Smith LM, Auble DT. Formaldehyde crosslinking: a tool for the study of chromatin complexes. J. Biol. Chem. 290(44), 26404–26411 (2015).
    • 14. Forsthoefel DJ, Park AE, Newmark PA. Stem cell-based growth, regeneration, and remodeling of the planarian intestine. Dev. Biol. 356(2), 445–459 (2011).
    • 15. Forsthoefel DJ, Waters FA, Newmark PA. Generation of cell type-specific monoclonal antibodies for the planarian and optimization of sample processing for immunolabeling. BMC Dev. Biol. 14(1), 45 (2014).
    • 16. Ross KG, Omuro KC, Taylor MR et al. Novel monoclonal antibodies to study tissue regeneration in planarians. BMC Dev. Biol. 15(1), 2 (2015).
    • 17. Newmark PA, Sánchez Alvarado A. Bromodeoxyuridine specifically labels the regenerative stem cells of planarians. Dev. Biol. 220(2), 142–153 (2000).
    • 18. Sánchez Alvarado A, Newmark PA. Double-stranded RNA specifically disrupts gene expression during planarian regeneration. Proc. Natl Acad. Sci. USA 96(9), 5049–5054 (1999).
    • 19. Gambino G, Iacopetti P, Guidi P et al. Cell quiescence in planarian stem cells, interplay between p53 and nutritional stimuli. Open Biol. 12(12), 220216 (2022).