Main

Multicellular organisms rely on individual cells to detect different stimuli and translate them into various biological responses. Correspondence must be maintained between an extracellular input and its specific cellular response output. Decades of research established chains of molecular events from ligand perception by cell surface receptors to transcriptional outputs in the nucleus. Yet a key question remains: how is pathway-specific information maintained all the way to the nucleus, when multiple receptor pathways are stimulated and converge on a limited number of common signalling intermediates? The mitogen-activated protein kinase (MPK) cascade is a key intermediate module, responding to numerous stimuli in plants, animals and fungi, leading to a diverse array of outputs1,2,3,4. Understanding the mechanisms underlying specificity in such an hourglass architecture presents a substantial challenge.

In plants, this ‘hourglass problem’ is amplified due to their cell types being more broadly competent, compared with highly specialized mammalian cells. Arabidopsis, for instance, displays over 200 leucine-rich-repeat receptor-like kinases (LRR-RLKs)5,6, dwarfing the 11 Toll-like receptors found in humans7, both representing a subset of their total receptor complement. At the same time, Arabidopsis has much less cell types than any mammal8,9, enhancing the potential complexity of receptor pathways on a per-cell basis. In plants, a prevailing notion is that common pathway components mediate specific outputs by activating different sets of transcription factors, expressed in different cell types. Yet, this would imply that a single cell-type would accommodate only one receptor/ligand pathway, which is implausible for plant cells. Therefore, it is crucial to understand how different receptors in a single plant cell maintain signalling specificity. However, such studies are scarce due to the challenges of isolating and analysing signalling events at single-cell resolution, as well as the lack of precise cellular readouts.

To address these challenges, we leveraged the ordered, linear differentiation of transparent Arabidopsis roots, allowing direct observation of single cells within an intact organism. Within this context, we established the Arabidopsis root endodermis as a cellular model to compare two well-studied LRR-RLK receptor pathways at single-cell level (Fig. 1a). One pathway uses the FLAGELLIN SENSING2 (FLS2) immune receptor, recognizing the microbial pattern flg22 (refs. 10,11). The other pathway uses the SCHENGEN3 (SGN3, also called GASSHO1 (GSO1)) receptor and its endogenous Casparian Strip Integrity Factor 1/2 (CIF1/2) ligands12,13. The endodermis is a specialized cell layer found in all vascular plants, forming a ring-like Casparian strip (CS), a highly localized membrane–cell wall nexus (Fig. 1d) analogous to tight junctions in animal intestinal epithelia14. This CS is composed of a lignified cell wall, creating an extracellular diffusion barrier in roots15,16. SGN3 enables the plant to detect developmental or stress-induced discontinuities in its CS, manifested as CIF peptide leakage from inner cell layers12,17. During endodermal differentiation, the SGN3 pathway drives CS maturation, characterized by fusion of its membrane domains. In situations of chronic or strong stimulation, the SGN3 pathway additionally induces ectopic lignification of cell corners and excess suberization (Fig. 1a)18,19,20. These precise and distinctive subcellular features of the SGN3 pathway were used as unambiguous and quantifiable signalling readouts.

Fig. 1: Endodermis-specific FLS2 induces ectopic lignin but cannot replace endogenous SGN3 for Casparian strip domain fusion.
figure 1

a, A single cell-type signalling system in the root endodermis, comparing the SGN3/CIF2 developmental pathway and FLS2/flg22 immunity pathway. b, Endodermis-specific expression and localization of SGN3 (pSGN3::SGN3-mVenus in wild type) and FLS2-GFP under pSGN3 or pCASP1 in fls2 or fls2 sgn3 backgrounds. Representative overviews (maximum projection) and zoomed surface views (see d) of mature CS root cells are shown. This experiment was repeated twice with similar results. Colour bar shows gradient intensity of GFP and mVenus (mV). c, CS and CASP1 domain fusion in 2-in-Endo CASP1-GFP after 1 d treatment with H2O, 100 nM CIF2 or flg22. Left: Untreated 2-in-Endo root scan outlined by calcofluor white staining of cellulosic cell walls (black). Endodermal cell surface views (see d) at mature CS stage (16th–17th endodermal cell after onset of elongation abbreviated 16th–17th cell) show CASP1-GFP (green), lignin stained with basic fuchsin (magenta) and overlaps of the two channels (Merged). Note that FLS2-GFP fluorescence is too weak to be detected with the same settings used to detect CASP1-GFP. Arrowheads highlight ectopic lignin. ‘Outer’, cortex-facing; ‘inner’, stele-facing endodermal surface. Scale bars, 5 μm. d, Schematics of CS development and three-dimensional architecture. Top left: Wild-type root shows CS maturation from disconnected domains to fused rings, functioning as a root apoplastic barrier. Early maturation occurs around the 11th–12th cell (mostly fused), while the 16th–17th cell reaches full maturity (fully fused). Right: SGN3 pathway mutants cannot fuse the CS domains and lack a functional barrier. Bottom: 2-in-Endo fuses the CS domains in response to CIF2, but not to flg22, yet both trigger ectopic lignin (black arrowheads) at the cortex side. Illustrations adapted from ref. 20 under a Creative Commons license CC BY 4.0. e, Quantifications for c showing the percentage CASP1-GFP gaps in 2-in-Endo in response to 1 d treatment with H2O, 100 nM and 1 µM CIF2 or flg22 (n ≥ 8 individual roots). f, PI penetration assay quantifies CS barrier function in 2-in-Endo after 1 d treatments compared to controls (n ≥ 10 individual roots). Roots with no barrier show PI penetration near the root–hypocotyl junction, shown in the ‘near hypocotyl’ category and are excluded from numerical statistical tests. For e and f, groups with the same letter are not significantly different (P < 0.05, one-way ANOVA and unbalanced Tukey’s test for unequal replication).

The SGN3 and FLS2 pathways are regarded to have markedly different functional outputs (a precise developmental programme versus a broad bacterial immune response), yet they display striking similarities in the downstream signalling components. Both receptors use the same small family of co-receptors to bind their respective ligands, flg22 and CIF2 (refs. 21,22,23). Both require members of the same small family of SERK co-receptors23,24 and subsequently activate members of the receptor-like cytoplasmic kinase VII family20,25, which triggers a series of common intracellular signalling events, including reactive oxygen species (ROS) production and activation of MPKs20,26,27,28,29. How are different outputs achieved when pathways share such extensive similarities? Narrowing the broad expression of FLS2 into the endodermal SGN3 domain sharpens the question of signalling specificity in plants, because it finally allows us to address what happens when two pathways are stimulated in a single cell-type.

Using our tailored genetic background, we provide clear evidence for the maintenance of signalling specificity in a single cell despite strong pathway activation. We identify the transcription factor MYB36, a master regulator of endodermal differentiation19,30, as a key hub driving pathway-specific outputs in the nucleus. Through extensive, cell-type-specific loss and gain-of-function studies of MPKs and MPK kinases (MKKs), we unveil their distinct capacities for transducing both common and specific branches of the two signalling pathways. We demonstrate that so-called ‘common intermediates’, such as MPK cascades, can produce a wide range of outputs through combinatorial activation of common and specific components.

Endodermis-specific FLS2 cannot replace endogenous SGN3 for CS domain fusion

To allow stimulation and direct comparison of two distinct pathways in the differentiating root endodermis (Fig. 1a), we expressed FLS2 under the endodermis-specific SGN3 promoter (pSGN3::FLS2-GFP) in an fls2 mutant background (Fig. 1b). We then knocked out CIF1 and CIF2 genes using CRISPR-Cas9, ensuring that SGN3 signalling only occurs upon external peptide stimulation. We termed the resulting plant line (pSGN3::FLS2-GFP fls2 cif1 cif2) as ‘2-in-Endo’. CIF2 peptide treatments on the cif1 cif2 mutant can efficiently restore the SGN3 pathway function and re-establish a continuous Casparian strip20,23; flg22 peptide treatments rapidly activate immune signalling in the endodermis from both native31 and endodermis-expressed FLS2 in the root31,32. Using this line, we investigated whether FLS2 could functionally replace SGN3.

SGN3 pathway stimulation causes growth and fusion of the Casparian strip domains (CSD) from aligned islands (called ‘string-of-pearls’ stage) into a continuous belt (Fig. 1d)12,13,33. Casparian strip domain protein 1 (CASP1) marks this domain and predicts lignin deposition at the CS26,34. We therefore introduced pCASP1::CASP1-GFP (CASP1-GFP) into 2-in-Endo. We confirmed that CIF2 treatment triggers full connection of CASP1 domains and corresponding CS lignin, leaving no gaps at a stage where the CS is fully mature in wild-type roots (Fig. 1c,d and Supplementary Video 1). In contrast, treatments with flg22 cannot improve CS domain fusion, showing comparable percentage of gap areas to controls (Fig. 1e). This was confirmed by visualizing CASP1-GFP from initiation to maturation (Extended Data Fig. 1a) and by quantifying discontinuity in overviews (Extended Data Fig. 1b,c).

Importantly, this distinct outcome is not due to a lack of FLS2 functionality, as flg22 treatment induces similar ROS production and ectopic lignification as CIF2 treatment (Fig. 1c and Extended Data Fig. 1f). These additional SGN3 pathway outcomes are also observed upon strong CIF2 stimulation12,20. A third outcome of strong CIF2 stimulation is precocious (nearer to the root tip) suberin deposition, also mimicked by flg22 treatment in 2-in-Endo (Fig. 1a and Extended Data Fig. 1d). Therefore, endodermis-specific FLS2 signalling is able to replicate some outcomes of SGN3, but is unable to mediate the crucial aspect of CASP and CS domain growth and fusion. Even with higher FLS2 receptor levels in an sgn3 background (pCASP1::FLS2-GFP fls2 sgn3, Fig. 1b), FLS2 stimulation cannot induce CASP domain fusion (Extended Data Fig. 1g,h).

This suggests that CS domain fusion is insensitive to FLS2 signalling, clearly indicating the presence of a main SGN3 signalling branch that FLS2 stimulation cannot induce.

Blocking of the propidium iodide (PI) fluorescent tracer provides a sensitive assay to measure CS functionality—it quantifies onset of an operational extracellular diffusion barrier by scoring PI exclusion from the inner vascular tissues (Extended Data Fig. 1e)15,16. In wild-type roots, blocking of PI uptake coincides with the formation of a continuous CS, assured by a functional SGN3 pathway (Fig. 1f). A delayed PI exclusion can still occur in the absence of a CS through SGN pathway-induced compensatory lignification at cell corners. This is observed in myb36, a CS-lacking mutant, but not in myb36 cif1 cif2 (Fig. 1f). Note that the PI block in myb36 is achieved purely by compensatory lignin, as the mutant lacks any CS lignin (Fig. 3b)19. This blockage is typically delayed by ~20 cells compared with wild-type plants with a CS (Fig. 1f).

We found that absence of a diffusion barrier in 2-in-Endo (H2O treatment) is fully rescued by CIF2 treatment, restoring a wild-type diffusion barrier (Fig. 1f and Extended Data Fig. 1e). By contrast, flg22 treatments activate delayed barrier formation in 2-in-Endo, similar to myb36, through ectopic lignin formation (Fig. 1f). In the overexpressing pCASP1::FLS2 line, the diffusion barrier is established earlier by flg22 (Extended Data Fig. 1i), probably due to stronger ectopic lignification (Extended Data Fig. 1g). Nevertheless, in both cases, barriers are still significantly delayed compared with CIF2-stimulated 2-in-Endo or wild-type lines.

Thus, despite being able to induce lignification and suberization in the endodermis, FLS2 signalling cannot functionally replace SGN3 for the growth and fusion of CASP1 and CS domains, indicating that signalling specificity between two similar receptor pathways is maintained within the same cell-type.

Endodermis-expressed FLS2 and SGN3 activate both common and pathway-specific transcriptional responses

To compare transcriptional outputs of FLS2 and SGN3 pathways solely within the endodermis, we conducted comparative RNA-seq analysis of pCASP1::FLS2 fls2 and wild-type seedling roots respectively treated with flg22 and CIF2 at three timepoints (30, 120, 480 min) (Extended Data Fig. 2a, CIF2 responses data from ref. 20). The receptor mutant controls sgn3 and fls2, and displayed minimal gene responses to respective peptides (Fig. 2a). We found a substantial overlap between the SGN3 and the FLS2 pathway, with 469 out of 1,262 differentially expressed (DE) genes in common (Fig. 2a). The most enriched Gene Ontology (GO) categories of both pathways show a high degree of overlap, reflecting general biotic and abiotic stress responses (Fig. 2b). The shared functional outcomes that we observed are reflected in the transcriptional profiles: genes involved in lignin and suberin biosynthesis show matching induction patterns after CIF2 or flg22 treatment in the respective backgrounds (Fig. 2c,d). This commonality cannot be simply attributed to shared cell identity (the endodermis), as a cross analysis of highly flg22-responsive genes in whole wild-type seedlings35 reveals mirrored induction patterns in the endodermis (Extended Data Fig. 2b).

Fig. 2: Endodermis-expressed FLS2 and SGN3 trigger common and pathway-specific transcriptional responses.
figure 2

a, Number of genes significantly regulated by CIF2 or flg22. Cut-off: |log2 fold change (FC)| > 1 and P < 0.05 for at least one timepoint. Genes that are significantly regulated by one pathway but not the other are considered specific. ‘WT.CIF2’, ‘sgn3.CIF2’: wild-type/sgn3 + 1 µM CIF2; ‘FLS2.flg22’, ‘fls2.flg22’: pCASP1::FLS2-GFP fls2/fls2 + 1 µM flg22. See Supplementary Fig. 2a for setup. b, GO-enrichment analysis of FLS2 and SGN3 pathways in endodermis. Colour: GO-enrichment significance. c,d, Heat maps of lignin (c) and suberin (d) biosynthesis-related gene expression fold changes post-peptide versus H2O treatment at indicated timepoints. e, Heat maps of fold changes for MYB36-regulated genes, significantly downregulated in myb36 compared with wild-type (P < 0.001). For ce, asterisks indicate significant regulation (P < 0.05). Colour bar: degree of fold change. f, Time-lapse images of pMLO3::NLS-3xmVenus (top) and pCYP715A1::NLS-3xmScarlet (bottom) transcriptional responses in 2-in-Endo to 1 µM CIF2 or flg22. Sum slice projections of roots at 3rd–7th cell (top) and 21st–25th cell (bottom) shown. Colour bar shows gradient intensity of mV or mScarlet (mS). Scale bars, 25 µm. Right: Quantifications show total grey intensity of nuclei signal at each hour (mean ± s.e., n = 6 individual roots).

Despite this overlap, we found 158 and 635 DE genes and GO terms specifically associated with SGN3 and FLS2 pathways, respectively (Fig. 2a and Extended Data Fig. 2d). The greater number of DE genes in the FLS2 pathway could arise from the stronger endodermal CASP1 promoter driving FLS2, compared with the native SGN3 promoter. To validate pathway-specific response genes (Extended Data Fig. 2c), we generated transcriptional fluorescent reporters in 2-in-Endo and found that the majority of reporter lines match the RNA-seq predictions (Extended Data Fig. 2c and Supplementary Table 1). For example, MILDEW RESISTANCE LOCUS O 3 (MLO3) shows a much stronger transcriptional response to CIF2 than to flg22 (Fig. 2f). Other SGN preferential reporter genes include PRK1, LAC3 and MCA2 (Supplementary Table 1 and Video 2). In contrast, the cytochrome P450 family gene CYP715A1 is an FLS2-specific reporter that responds strongly to flg22 but is non-responsive to CIF2 (Fig. 2f), with other examples including WRKY30, LAC1 and PER52 (Supplementary Table 1 and Video 3). Some reporters respond equally well to either flg22 or CIF2 in 2-in-Endo, with many showing non-cell-autonomous responses in surrounding root cell-types (for example, WRKY41, PER71 and ERF105) (Supplementary Table 1).

Intriguingly, our dataset revealed that the top 10% most responsive genes (by fold change) in early timepoints are common to both pathways, whereas genes responding more weakly (but significantly) exhibited more pathway specificity (Extended Data Fig. 2e). We speculate that ‘fast and strong’ responses are often regulated by the common signalling modules of the two pathways.

The transcription factor MYB36 controls SGN3-specific transcriptional responses

Our transcriptional analyses suggested that the transcription factor MYB36, reported to be a key regulator of endodermis differentiation19,30, also acts as a hub to regulate SGN3-specific responses. We found that genes regulated by MYB36 (downregulated in myb36 compared with wild type30) tend to respond specifically to CIF2 (Fig. 2e). While the scale of induction by CIF2 is relatively weak (log2 FC < 2), many of these genes are specifically expressed in the endodermis and are known to contribute to CS integrity. For example, expression of CASP genes (CASP1/2/3/4) is crucial for establishing a functional CSD34,36, while peroxidases such as PER9 and PER64 mediate CS lignification37, and the dirigent protein ESB1 is essential for both CSD formation and lignification38. These genes might therefore carry out SGN3-specific functions and their non-responsiveness to flg22 can straightforwardly explain the inability of flg22 to promote CS domain fusion.

CIF2-induced CASP domain fusion requires de novo protein synthesis20. We hypothesize that MYB36-mediated transcriptional responses upon SGN3 stimulation enable CASP domain fusion, while the FLS2 pathway is incapable of activating MYB36. Consequently, the SGN3 pathway would function independently of MYB36 for ectopic lignification but require MYB36 for CASP and CS domain fusion. MYB36 remains partially functional in sgn3 and cif1 cif2, but only mediates formation of aligned CASP domain islands. The role of SGN3 would therefore be to drive domain fusion by boosting the activity of MYB36.

Predicted MPK phospho-sites in MYB36 are important for CS domain fusion

MYB36 was shown to be phosphorylated by specific MPKs (MPKs 2, 3 and 16) in an in vitro protein microarray39. Three MPK phosphorylation motifs (serine proline) are predicted in MYB36 (Extended Data Fig. 3a). To test the potential necessity and sufficiency of MYB36 phosphorylation at these sites for CS domain fusion, we mutated all three serines to generate a phospho-null variant, MYB36AAA and a phosphomimic variant, MYB36DDD.

The untagged MYB36AAA, introduced in myb36 mutant, can restore CASP1 expression and accumulation in the endodermis, but often failed to complement CASP1 domain fusion (Fig. 3a and Supplementary Table 2). This suggests that MYB36AAA, while active, is less functional than either the wild-type MYB36WT or the phosphomimic MYB36DDD, both of which fully complement CASP1 domain fusion (Fig. 3a and Supplementary Table 2). The fusion defects exhibited by MYB36AAA can be described as a band of ‘speckles’ roughly aligned with the endodermis central plane (Fig. 3b). These CASP1 speckles consist of many smaller disconnected patches, in comparison with the larger, more connected domains caused by the loss of SGN3 signalling in sgn3 or cif1 cif2 (Fig. 3a–c and Extended Data Fig. 3c). As expected, the ability to fuse CASP1 domains correlates with the formation of a continuous CS lignin (Fig. 3b) and early formation of a diffusion barrier (Fig. 3d).

Fig. 3: Predicted phospho-sites of MYB36 important for SGN3-dependent CS domain fusion.
figure 3

a, CASP1-GFP (gradient intensity) in myb36 CASP1-GFP transformed with untagged MYB36 genomic DNA constructs: WT, MYB36AAA (S18A, S146A, S169A), MYB36DDD (S18D, S146D, S169D). T2 roots at mature CS stage (16th–17th cell) are shown. Untransformed myb36, wild type and sgn3 with CASP1-GFP are shown as controls. For T1 phenotype distribution, see Supplementary Table 2. Colour bar shows gradient intensity of GFP. b, CS and CASP1 domain fusion in myb36 transformed with untagged MYB36 variants after 1 d of 1 µM CIF2 or H2O treatment. Endodermal cell surface views at mature CS stage (16th–17th cell) show CASP1-GFP (C, green), lignin (L, magenta) and overlaps of the two channels (Merged). Arrowheads show ectopic lignin on cortex-facing side. Scale bars, 5 μm. c, Quantification of CASP1 domain properties: number of CASP1-GFP fragments per 100 µm in surface views. myb36 CASP1-GFP transformed with untagged MYB36WT (WT), MYB36AAA (AAA) or MYB36DDD (DDD) are treated for 1 d with H2O (grey) and 1 µM CIF2 (pink) (n ≥ 10). For fragment size, see Supplementary Fig. 3c. d, PI penetration assay quantifies CS barrier function of myb36 CASP1-GFP transformed with MYB36 constructs (WT, AAA, DDD), untagged or with C-terminal mCherry or Turbo-GFP tag (short for 6xHis3xFLAG-TurboID-GFP) (T1 roots, n ≥ 10). Multiple comparisons were done separately for each tag group, always including 2 controls and 3 lines per tag group. e, Quantification of CS discontinuity of sgn3 CASP1-GFP transformed with untagged MYB36WT, MYB36AAA or MYB36DDD (T1 and untransformed; n ≥ 8). f, Schematic of working models. SGN3 signalling acts through MYB36, possibly by boosting phosphorylation, to promote CASP domain fusion. Ectopic lignin is induced by SGN3 signalling independently of MYB36; MYB36AAA is sufficiently active to induce CASP domain islands. MYB36AAA cannot mediate CIF2-stimulated domain fusion but can mediate CIF2-stimulated ectopic lignin and acts via an unknown transcription factor (TFx); MYB36DDD is able to enhance CASP domain fusion without the SGN3 pathway. All data from roots at mature CS stage (16th–17th cell). For ce, groups with the same letter are not significantly different (P < 0.05, one-way ANOVA and Tukey’s HSD test). Untrans, untransformed.

Importantly, we found that the domain fusion defect in myb36 MYB36AAA is resistant to external CIF2 application (Fig. 3b,c and Extended Data Fig. 3c,d), suggesting that CIF2-induced domain fusion is impaired by the absence of functional phospho-sites in MYB36 (Fig. 3f middle). By contrast, CIF2-induced ectopic lignification is unaffected by MYB36AAA, since ectopic lignin in myb36 MYB36AAA is further enhanced by external CIF2 application (Fig. 3b). Accordingly, myb36 MYB36AAA has a delayed PI block, similar to untransformed myb36 (Fig. 3d).

Our data strongly suggest that the SGN3 pathway operates through multiple branches, one requiring MYB36 activation for CASP domain fusion, probably via MPK phosphorylation, and another triggering compensatory ectopic lignification independent of MYB36 (Fig. 3f left). Specificity could thus arise from the FLS2 pathway’s inability to activate MYB36.

We also found that different C-terminal tags affect the functionality of MYB36AAA, with large tags such as Turbo-GFP (847 aa) and mCherry (406 aa) apparently enhancing functionality of all MYB36 variants compared with their untagged version (Fig. 3d and Supplementary Table 2), possibly due to enhanced stability. Nevertheless, despite similar accumulation levels, MYB36AAA-mCherry was still unable to fully complement CASP domain fusion in myb36, unlike the other variants (Extended Data Fig. 3b).

The phosphomimic MYB36DDD could fully complement CASP domain fusion and CS barrier function in myb36, comparable to MYB36WT (Fig. 3a–d). Interestingly, the untagged MYB36DDD shows gain-of-function activity, improving CASP domain fusion in sgn3, whereas MYB36AAA could not (Fig. 3e,f right). This further supports the idea that the predicted MPK phospho-sites in MYB36 are essential for CASP domain fusion induced by the SGN3 pathway. Furthermore, MYB36 protein accumulation and stability appear to be important determinants of its activity within the SGN3 pathway.

Multiple MPKs contribute to CS integrity in the SGN3 pathway

MPKs are direct, key regulators of transcription factors, and our data indicate their potential in directly regulating MYB36 activity. Yet, several MPKs are reported to be activated by both SGN3 (ref. 20) and FLS2 pathways27,40. In 2-in-Endo seedling roots, we confirmed that both pathways can activate MPK3 and MPK6 in the endodermis upon flg22 or CIF2 treatments (Fig. 4a). To obtain a more global picture of MPK activities in the endodermis, we generated an expression atlas for all 20 MPKs in Arabidopsis (Supplementary Table 3) by generating and analysing individual transcriptional fluorescent marker lines. Overall, we observed broad expression patterns across different tissues (Supplementary Table 3). We found that at least 12 MPKs are expressed in the endodermis, hence a subset could easily transduce specific signals downstream of SGN3 and FLS2 (Extended Data Fig. 4b,c). Integrating our atlas with RNA-seq30 and translating ribosomal affinity purification (TRAP) data enriched in the endodermal cell file (Extended Data Fig. 4a), we homed in on 8 candidates (MPK2/3/6/9/15/16/17/19). However, none of the tested mpk single mutants show strong defects in CS barrier function or CASP1 domain fusion (Fig. 4b and Extended Data Fig. 5a). We nevertheless observed a slight delay in forming a diffusion barrier in mpk2 (Fig. 4b), fitting with the in vitro phosphorylation data of MYB36 being a substrate for MPK2, 3 and 16 (ref. 39). However, no obvious discontinuities persist at maturity in mpk2 CASP1-GFP (Extended Data Fig. 5a). Moreover, neither T-DNA nor CRISPR mpk2 mpk16 double mutants showed a significant barrier diffusion or CASP1-GFP defect (Fig. 4b and Extended Data Fig. 5a,c), suggesting that higher-order mutants will be required for observing strong and reliable CS formation phenotypes.

Fig. 4: Multiple MPKs contribute to CS integrity in the SGN3 pathway.
figure 4

a, Immunoblots (IB) against p42/44 (phosphorylated MAPKs) indicate sizes corresponding to phosphorylated MPK6 (top arrowhead) and MPK3 (bottom arrowhead). 2-in-Endo seedling roots treated with H2O, 1 µM CIF2 or flg22 for 5, 10 and 15 min. Loading controls: IBs with anti-MPK6 antibody. See Source Data Supplementary Fig. 7a for original blots, Source Data Supplementary Fig. 7b for IBs on whole seedlings of 2-in-Endo with Ponceau S as loading control and Source Data Supplementary Fig. 7c with anti-MPK6 with mpk6 as negative control. This experiment was repeated twice with similar results. b, PI penetration assay quantifies CS barrier function in various mpk T-DNA mutants and mpk2 mpk6 CRISPR mutants (10 ≤ n ≤ 49) compared to wild type (n = 73) and sgn3 (n = 10). Data from multiple independent experiments are combined. Unbalanced Tukey’s HSD test for unequal replication. See Extended Data Fig. 5c for allele description of the two CRISPR mutant lines. c, PI penetration assay with endodermal suppression of multiple MPK activities by inducing HopAI1 vs inactive HopAIH103A in wild type (n ≥ 6). Endodermis-specific estradiol (EST) inducible promoter: pLTPG15::XVE»pLexA::HopAI1 (indHopAI1). d, PI penetration assay upon conditional loss of function of both MPK3 and MPK6 (n ≥ 10). pMPK6::MPK6YG or pMPK3::MPK3TG are sensitive to NM-PP1 in mpk3 mpk6 background. Seedling roots (4-day-old) transferred to 5 µM NM-PP1 or DMSO for 1 d. e, Quantification of CS discontinuity upon endodermal induction of constitutively active MPK6a or MPK2a in sgn3 CASP1-GFP (n ≥ 8). f, PI penetration assay upon endodermal induction of MPK6a or MPK2a in sgn3 CASP1-GFP. Two-sided Welch’s t-test compares indMPK6a (n = 12) to the wild type (n = 10), ***P = 5.72 × 10−14. g, CS lignin and CASP1 phenotypes upon endodermal induction of MPK6a or MPK2a in sgn3 CASP1-GFP at 16th–17th and 30th endodermal cells after onset of elongation. Surface views show CASP1-GFP (C, green) and lignin (L, magenta). Arrowheads show ectopic lignin on cortex-facing side. Scale bars, 5 μm. For be, groups with the same letter are not significantly different (P < 0.05, one-way ANOVA and Tukey’s HSD). ‘ind’ designates ‘pLTPG15::XVE»pLexA::’. For EST induction, all seedlings were grown for 5 d on 5 µM EST or DMSO non-induced control before analysis. Whiskers indicate maximum and minimum values, except outliers (defined as >1.5× interquartile range above 3rd quartile or below 1st quartile, respectively).

Because MPKs are involved in many processes, dissecting their function without pleiotropic effects in higher-order mutants is challenging. To circumvent this problem, several alternative tools were tested. First, we expressed HopAI1, a Pseudomonas syringae effector that specifically deactivates multiple MPKs, including MPK3/4/6 (refs. 41,42,43), under an endodermis-specific estradiol (EST) inducible promoter (pLTPG15::XVE>>pLexA::HopAI1, hereafter indHopAI1). HopAI1 induction in wild type leads to delayed barrier formation and some ectopic lignin, suggesting that MPK activities are required for CS functionality. No phenotype is observed using an inactive HopAI1H103A mutant (Fig. 4c and Extended Data Fig. 4d). However, at our confocal resolution, HopAI1 induction does not seem to affect CASP domain fusion (Extended Data Fig. 4d).

Since mpk3 mpk6 double mutants are embryo lethal44, we used conditional loss-of-function lines expressing pMPK3::MPK3TG or pMPK6::MPK6YG in an mpk3 mpk6 background45,46. MPK3TG and MPK6YG are specifically sensitive to NM-PP1, a bulky analogue of the general kinase inhibitor PP1. Indeed, 1-day inhibitor treatment of these lines delayed the PI block without causing other developmental defects (Fig. 4d).

We also used a gain-of-function approach to investigate whether constitutively active MPK6a and MPK2a can alleviate CS defects in sgn3. We found that expressing MPK6a or MPK2a under an inducible endodermis-specific promoter (ind) in sgn3 reduces CASP1 domain discontinuities at maturity (Fig. 4e). Interestingly, only induction of MPK6a, but not MPK2a, partially restores the diffusion barrier in sgn3 (Fig. 4f and Extended Data Fig. 5b). This correlates with MPK6a activating ectopic ROS and ectopic lignin and suberin formation in sgn3, in contrast to MPK2a (Fig. 4g, and Extended Data Figs. 4e,f, and 7d). MPK6a thus appears to have a broader activity, affecting specific and common branches of the SGN3 pathway, while MPK2a contributes more specifically to CASP domain fusion.

Since MPK6a is sufficient to induce ectopic lignin in sgn3, we investigated whether MPK6 is necessary for SGN3-dependent ectopic lignification by testing whether CIF2-induced ectopic lignin is affected in mpk6. We found that 1-day CIF2 treatment could induce similarly strong ectopic lignin in mpk6 and wild type, as well as in mpk3 (Extended Data Fig. 4g). This suggests that genes in addition to MPK6 must contribute to ectopic lignification downstream of the SGN3 pathway. Despite MPK2a and MPK6a contributing to domain growth and fusion, neither alone is sufficient to complete CS fusion. Therefore, a combination of different MPK activities appears to contribute to the three-branched outputs of the SGN3 pathway, some unique and some overlapping with the FLS2 pathway.

Activation of individual MKKs in the endodermis produces distinct outputs

Arabidopsis contains 80 MAPKKKs and 20 MAPKs, but only 10 MKKs, the direct upstream regulators of MPKs47,48. MKKs thus represent the narrowest part of the hourglass and the most critical step for information transfer. MKKs tend to selectively activate certain MPK combinations39,49, suggesting that the activation of different MKKs may translate into distinct combinations of MPK activities, depending on the specific pathway triggered.

Given the relatively small number of MKKs, it is feasible to conduct a comprehensive examination of their activities in the endodermis. Since we did not find even weak phenotypes in any of the single and double mutants tested (Extended Data Fig. 7e,f,g), we decided to use our endodermal-specific induction system and introduced all 10 constitutively active MKKs in the sgn3 background (designated indMKK1a–indMKK10a). The 10 MKKs are classified into phylogenetic groups (A–D)47 (Fig. 5a) and interestingly, these groupings match strikingly well with the functional outcomes we observe in the endodermis.

Fig. 5: Activation of individual MKKs in the endodermis produces distinct outputs.
figure 5

a, MKKs 1–10 from Arabidopsis thaliana are classified into 4 phylogenetic groups using full length sequence47. b, CASP1 and lignin phenotypes upon endodermal induction of constitutively active MKK1–10a in sgn3 CASP1-GFP at 16th–17th cell. Endodermal cell surface views show CASP1-GFP (green) and lignin (magenta). Arrowheads show ectopic lignin on cortex-facing side. Scale bars, 5 μm. c, Quantification of CS continuity upon endodermal induction of MKK1–10a in sgn3 CASP1-GFP (n ≥ 11). d, Quantification of CS coverage upon induction of MKK7a and MKK9a in wild type and sgn3 CASP1-GFP (n ≥ 10). At mature CS stage, total CASP1 length of each overview image measured normalized to the average CASP1-GFP length in wild type to calculate percentage of CS coverage for each data point. e, PI penetration assay quantifies CS barrier function upon endodermal induction of MKK1–10a in sgn3 CASP1-GFP (n ≥ 10). f, PI penetration assay upon endodermal induction of MKK9a in wild-type CASP1-GFP and in sgn3 CASP1-GFP (n ≥ 10). See Extended Data Fig. 6f for T1 data and statistics. For ce, two-sided Welch’s t-test compares DMSO with EST for each genotype, P values were adjusted for multiple comparisons by Bonferroni correction (***P < 0.001, **P < 0.01, *P < 0.05). For EST induction, all seedlings were grown for 5 d on 5 µM EST or DMSO. For each indMKKa genotype, two independent T2-line data are combined in cf. Whiskers indicate maximum and minimum values, except outliers (defined as >1.5× interquartile range above 3rd quartile or below 1st quartile, respectively). Ctrls, controls.

For Group A/B members, namely MKK1/2/6/3, we found that induction of each individual MKKa in the sgn3 endodermis can improve CASP1 and CS lignin fusion (Fig. 5b,c), although none is sufficient to rescue the barrier defects of sgn3 in the diffusion assay (Fig. 5e). This can be attributed to the persistence of small gaps in the CS after induction (Fig. 5b), coupled with the fact that these activated variants do not induce compensatory ectopic lignin or ROS (Fig. 5b and Extended Data Fig. 7c,d). Intriguingly, unlike MPK6a, which triggers early excess suberin, MKK1/2/3a induction suppresses suberization in sgn3 roots (Extended Data Fig. 7a). Our findings suggest that MKK1/2/3/6 are involved in transducing the SGN3-specific branch of CS domain fusion, which aligns nicely with in vitro data of MKK1-MPK2 and MKK2-MPK2 phosphorylating MYB36 (ref. 39). This fits our hypothetical model, whereby distinct SGN3 and FLS2 outputs are defined by differential regulation of MYB36 via specific MKK-MPK signalling routes (Fig. 7)46.

Group C members MKK4 and MKK5 show broader activities. Induction of MKK4a can moderately improve domain fusion in sgn3 (Fig. 5c) but uniquely promotes ectopic lignin and ROS production, even in the absence of SGN3 signalling (Fig. 5b and Extended Data Fig. 7c,d). This ectopic lignin is sufficient to form a delayed diffusion barrier in contrast to no barrier with non-induced control in indMKK4a sgn3 (Fig. 5e). In addition, indMKK4a triggers early excess suberin accumulation (fluorol yellow staining) and suberin synthesis (GPAT5 expression marker) in sgn3 (Extended Data Fig. 7a,b). These data suggest that MKK4a mediates the compensatory branches of the SGN3 pathway (ectopic lignin and excess suberin), a function akin to MPK6a’s effects. Given that MKK4-MPK6 is activated by FLS2 for immune function27,50, this cascade could regulate the common downstream outputs of both SGN3 and FLS2 in the endodermis (Fig. 7). MKK5, often considered functionally redundantly with MKK4 in immune signalling27, has little effect on CASP1 domain fusion or ectopic lignification (Fig. 5b,c,e) but does slightly enhance suberin synthesis upon induction in the sgn3 endodermis (Extended Data Fig. 7a,b).

Among Group D members, MKK9 stands out as we found that the others (MKK7/8/10) are either absent or only weakly expressed in the root endodermis (Extended Data Fig. 6a)30. Strikingly, EST induction of MKK7a and MKK9a in sgn3 suppresses both CASP1 and CS lignin accumulation (Fig. 5b,d and Extended Data Fig. 6b,e). Electron microscopy confirms that MKK9a also abolishes ROS accumulation and membrane attachment, typical of a functional CS structure (Extended Data Fig. 7c). This suppression remains prominent in wild-type background with SGN3 present (Fig. 5d and Extended Data Fig. 6c). In wild type, MKK9a suppresses CASP1 accumulation and domain fusion but does not suppress ectopic lignification (Extended Data Fig. 6c), explaining the partial PI block phenotype (Fig. 5f and Extended Data Fig. 6f). MKK9a also has a potent inhibitory effect on suberin in sgn3 (Extended Data Fig. 7b,c), consistent with MKK9’s recognized role in ethylene signalling51,52 and the known inhibitory effect of ethylene activation on suberin in the endodermis53. The other Group D members, MKK8a and MKK10a, improve CASP1 domain fusion, but not diffusion barrier defects, resembling the action of Group A/B members (Extended Data Fig. 5c,e)54,55. Western blot analysis confirms the enhanced accumulations of MKK1–10a upon EST induction (Extended Data Fig. 6d).

In summary, our systematic analysis of all 10 MKKs in a single cell-type demonstrates that activation of individual MKKs in the endodermis leads to clearly distinct output patterns. These patterns match both the common and specific outputs upon SGN3 and FLS2 stimulation in the endodermis. The Groups A/B (MKK1/2/3/6) are strong positive regulators specific to domain fusion; Group C MKK4 is the only one capable of promoting ectopic lignin and excess suberin. Finally, among Group D, MKK7 and MKK9 are strong negative regulators capable of suppressing CS development.

Constitutively active MKK9 inhibits CS formation via suppression of MYB36

The specific impact of MKK9 activation on CASP1 accumulation and fusion suggests that it could be a powerful suppressor of MYB36 activity. We tested this by introducing MYB36WT, MYB36AAA or MYB36DDD tagged with GFP-TurboID (here labelled -GFP) into the sgn3 indMKK9a background (Fig. 3d and Supplementary Table 2). With MYB36WT-GFP or MYB36AAA-GFP, indMKK9a still suppresses CASP1 accumulation in sgn3 (Fig. 6a,b). The phosphomimic MYB36DDD-GFP, by contrast, abolishes the inhibitory effect of indMKK9a on CASP1 accumulation (Fig. 6a,b). This indicates that MKK9 suppression is conditioned by the phosphorylation status of MYB36.

Fig. 6: Constitutively active MKK9 inhibits CS formation via suppressing MYB36.
figure 6

a, CASP1-GFP phenotypes of sgn3 CASP1-GFP indMKK9a (T3) transformed with Turbo-GFP-tagged MYB36WT-GFP, MYB36AAA-GFP and MYB36DDD-GFP. T2 roots at mature CS stage (16th–17th cell) are shown. b, Quantification of CS coverage upon induction of MKK9a sgn3 CASP1-GFP transformed with MYB36-GFP variants (T2, n ≥ 8). Groups with the same letter are not significantly different (P < 0.05, one-way ANOVA and Tukey’s HSD). c, Stability of MYB36 variants in sgn3 CASP1-GFP indMKK9a background with or without EST induction. Nuclei signals of MYB36-GFP at 7th–8th cell (start of CASP1-GFP deposition) corresponding to a are shown. CASP1-GFP and MYB36-GFP signals can be distinguished on the basis of different subcellular location. Colour bar shows gradient density of GFP. d, Quantification of MYB36 variants stability as in c, showing total grey intensity of nuclei signal (MYB36-GFP fluorescence) with or without EST induction at 7th–8th cell (T2, n ≥ 15). Two-sided Welch’s t-tests compare EST and DMSO for each genotype (***P ≤ 0.001, **P ≤ 0.01). For EST induction, all seedlings were grown for 5 d on 5 µM EST or DMSO before analysis. e, Quantification of CS continuity of 2-in-Endo CASP1-GFP upon 30 min, 1 µM flg22 pre-treatment plus 6 h, 100 nM CIF2 treatment or 100 nM CIF2 + 1 µM flg22 co-treatment ('Pre + Treatment', n ≥ 10). H2O treatments were used as controls. Groups with the same letter are not significantly different (P < 0.05, one-way ANOVA and Tukey’s HSD). For b, d, e boxplots, the whiskers indicate maximum and minimum values, except outliers (defined as >1.5× interquartile range above 3rd quartile or below 1st quartile, respectively).

We found that MKK9a induction reduces accumulation of MYB36WT-GFP and MYB36AAA-GFP in endodermal nuclei (Fig. 6c,d), and MYB36 levels correspond closely to the extent of CS suppression (Fig. 6a,c and Extended Data Fig. 8a). Conversely, MYB36DDD-GFP levels were unaffected by MKK9a induction, and MYB36DDD-GFP levels positively correlated with the rescue activity (Fig. 6 and Extended Data Fig. 8a,b). MKK9 signalling thus appears to destabilize wild type and the phospho-null variant of MYB36 to suppress its function. Intriguingly, in sgn3 indMKK9a lines expressing high levels of MYB36DDD-GFP, we could observe ectopic CASP1 patches outside of the CSD (Extended Data Fig. 8a), mimicking hyperstimulation of the SGN3 pathway12. We conclude that the phosphomimic variant of MYB36 not only has an ability to complement the sgn3 phenotype, but also reiterates SGN3 hyperstimulation, firmly placing phosphorylated MYB36 as an important downstream player of the SGN3 pathway.

The FLS2 pathway intertwines closely with ethylene signalling54,55,56 known to activate MKK1/3/951,52,57, and MKK7 was found to contribute to flg22-induced ROS burst58. Hypothetically, an activation of MKK9 (and/or MKK7) by the FLS2 pathway could account for the inability of flg22 stimulation to induce closure of CS domains by negatively regulating MYB36. This negative regulation could influence the activation from positive regulators of MYB36 activity and thus exacerbate the difference between SGN3 and FLS2 signalling outputs. If this were the case, flg22-FLS2 signalling should be able to interfere with CIF2-SGN3 signalling when simultaneously activated in the same cell. Indeed, we found that pre/co-treatment of flg22 with CIF2 leads to suppression of CIF2-induced CASP1 domain fusion in 2-in-Endo (Fig. 6e), lending support to such a model (Fig. 7b).

Fig. 7: Hypothetical model of two receptor pathways in a single cell-type.
figure 7

a,b, Hypothetical model of mechanisms underlying specificity of SGN3 signalling (a) and FLS2 signalling (b) in the endodermis based on current understanding.

Discussion

Signalling specificity is maintained at single-cell level

The complexity and intricacy of cellular signalling are increasingly recognized in plants59,60. Understanding the signalling dynamics at single-cell level is fundamental for delineating overall organismal responses. To enable both stimulation and observation of pathway activities at single-cell resolution, we utilized the unique characteristics of the SGN3 pathway, allowing for precise and quantifiable readouts in a non-dividing, easily observable cell.

Our study underlines the importance of examining signalling specificity beyond the constraints of tissue specificity and compartmentalization. We demonstrated that the FLS2 pathway in the endodermis could not substitute the SGN3 pathway in establishing a continuous Casparian strip (Fig. 1). This pertains to endogenous signalling, since the endodermis has the inherent capacity for FLS2 immune signalling31.

SGN3 as a neo-functionalized immune receptor?

A simplistic approach to achieve specificity at single-cell level would be to have dedicated sets of receptors, signalling components and transcription factors that are insulated between one pathway and another in the same cell. However, evolution appears to have favoured a more elegant solution—reusing key signalling components from diverse processes, a common theme among multicellular organisms61,62.

In plants, despite the expansion of the receptor repertoire for detecting a growing number of specific inputs, intracellular signalling pathways share striking similarities in their structure and components. It is thus reasonable to believe that outputs of many pathways share a common core. This is supported by the extensive interconnections among plant hormonal pathways63 and the substantial overlaps observed in transcriptomic responses to biotic and abiotic stresses64,65,66. Our data expand this idea and show that stimulation of a developmental and an immunity receptor kinase pathway can induce common ROS, lignin and suberin production (Fig. 1). This is corroborated by transcriptional profiles, showing matching induction patterns for genes involved in lignin and suberin biosynthesis following either CIF2 or flg22 treatments (Fig. 2). These commonalities raise an intriguing question: Could both pathways have originated from a shared signalling pathway module, having diverged over time to perceive distinct inputs while maintaining partially overlapping outputs? This fits the prevalent view of plant evolution being driven by cycles of gene duplication and neo-functionalization67. Once plants developed a mechanism for ROS production and lignification during defence, they may have adapted these responses for more precise developmental purposes, leading to the emergence of the primary function of the present-day SGN3 pathway, possibly through initially subtle divergences in the substrate specificity of the receptor kinase domains.

Intriguingly, our comparative RNA-seq analysis revealed substantial overlaps among early, highly responsive genes between the two pathways. Yet, despite this remarkable overlap, the FLS2 immune receptor kinase cannot replicate the central developmental function of the SGN3 pathway.

Combinatorial activation of MPK cascades can encode specific signalling outcomes

There is a remarkable potential for the combinatorial activation of kinases to encode signalling specificity in plants. Beyond the members of the MPK cascade, other extended kinase families, including several families of calcium-dependent kinases (CDKs)68, also transduce signals from LRR-RLKs. Our study focuses on the MPK cascade’s potential to confer a basic level of specificity, yet we acknowledge the role of additional signalling elements such as CDKs and phospholipids in contributing to the overall output specificity. Intriguingly, plant MPK cascades by themselves already exhibit a complex web-like structure, contrasting with the simpler, more linear pathways found in yeast and mammals62,69,70. Their expansion in plants has enhanced their capacity to form numerous combinations of MPKKK-MKK-MPK49,62. While our understanding of plant MPK network interactions has been advanced through global analyses and in vitro methods39,71,72,73, the mechanisms controlling specificity within these cascades remain largely unknown. MPK3 and MPK6, for example, are reported to be activated by numerous signalling pathways associated with diverse cellular functions4,74. Similarly, MPK1/2/7/14 are activated by various stress pathways75,76. This makes it challenging to understand how MPKs would mediate specific cellular responses. Our reporter expression atlas (Supplementary Table 3) and previous tissue-wide expression analyses indicate that MPKs are broadly expressed across tissues77,78, providing potential for combinatorial action. Our 2-in-Endo line ensures that downstream MPKs activated by both FLS2 and SGN3 pathways encounter identical sets of target proteins. This allowed us to assign the specific activity of SGN3 to its differential activation of MYB36, shown to be an MPK target in vitro39 (Fig. 3). Other endodermis-expressed MYB transcription factors, such as MYB15 and MYB41 respectively involved in lignin and suberin production79,80, are also reported to be phosphorylated by MPKs81,82 and might be mediators of the pathway responses common to FLS2 and SGN3, that is, ectopic lignin and excess suberin production (Fig. 7).

In plants, MKK–MPK interactions are known to be promiscuous, which might be highly relevant for generating signalling specificity39,49,71. Each MKK can generate a potentially unique pattern of MPK activation; each MPK could receive inputs from different MKKs depending on the pathway involved; and individual MPKs could activate both common and unique substrates83, suggesting an extensive interaction network. We propose that such combinatorial MPK activation patterns generate distinct ‘flavours’ that can drive specific outputs.

Elegant tissue-specific manipulations of MKKs in the stomatal lineage have demonstrated overlapping roles for MKKs 4/5 and 7/9 during early stages, and unique activities of MKK7/9 in late stages of stomatal development84,85,86. In addition, MKK4/5, but not MKK1/2/3/8/9, contribute to root meristem development87. These analyses revealed that MKKs have divergent functions over time along a complex developmental trajectory. Our simpler endodermal system uses a differentiating, non-dividing cell-type that allows distinct receptor stimulation and readouts at the same developmental stage. In our system, analysis of all ten MKKs provides compelling evidence that each MKK can activate distinct outputs within the same cell-type (Fig. 5). These functional groupings align well with MKK phylogeny and can account for both the common and unique outputs of the SGN3 and FLS2 pathways (Fig. 7).

On the basis of the matching readouts, we propose that the SGN3 pathway activates MKK1/2/3/6 (Groups A/B) to ensure domain fusion during CS development while suppressing excess suberin. Only upon strong or prolonged stimulation would the SGN3 pathway boost MKK4/5 (Group C) activity to promote ectopic lignin and excess suberin. By avoiding activation of negative regulators of CS development such as MKK9, the SGN3 pathway might further promote effective MYB36 activation. Among all MKKs, only MKK4 could enhance ROS production and lignification, consistent with a reported role of MKK4 in promoting ROS88. ROS homoeostasis could be achieved by balancing the activities of different MKKs downstream of SGN3, favouring CS domain closure upon low and transient stimulation, and preventing pleiotropic ectopic lignin and suberin accumulation until stronger, persistent stimulations are reached.

The FLS2 pathway in the endodermis, by contrast, might have a weaker ability to activate MKK1/2/3/6 (Groups A/B) but more potently activates MKK4/5 (Group C), leading to enhanced ectopic lignin and suberin. In addition, the inability of FLS2 to activate MYB36 may be reinforced by a stronger activation of MKK7/9, which would actively suppress MYB36 activity, fitting with the observed inhibitory effect of flg22 on CIF2 action (Fig. 6e). Previous analyses of the FLS2 pathway have painted much more complex and confusing pictures, with at least two independent MPK cascades, MKKK3/5-MKK4/5-MPK3/6 and MEKK1-MKK1/2-MPK427,29,89,90. Parts of this were found to be due to perturbations of guarding resistance proteins43,91,92,93.

We conclude that single cell-type analyses performed in a whole-tissue context can offer much clearer insights into signalling pathways. The complexities observed in previous studies may arise from readouts relying on protoplast experiments and/or use of total plant tissues. It is also intriguing to speculate that cells might switch between signalling states, with strong stimulation of one pathway actively suppressing another. Such dynamic modulation would allow a cell to respond to diverse stimuli, although not necessarily simultaneously.

Limitations of the study

This study provides new and conclusive evidence of divergent functional outputs of ten individual MKKs and two MPKs in a single cell-type, using a gain-of-function approach. However, future efforts require complementary loss-of-function analyses to genetically elucidate the MKK-MPK routes downstream of each pathway that lead to these distinct outputs. The major barrier is the redundancies and pleiotropic effects that arise from broad perturbation of multiple MPKs and/or MKKs. Potential workarounds, such as cell-type-specific MPK inhibition by HopAI1 phosphothreonine lyase activity, appears to be restricted to a subset of MPKs41,42,43. Future studies would benefit from testing additional approaches for precise tissue-specific MPK knockouts.

Our data support a compelling model of possible differential phosphorylation of MYB36 downstream of FLS2 and SGN3 pathways, driving output specificity. However, we have been unable to obtain sufficient amounts of MYB36 from 2-in-Endo to determine their phosphorylation states after peptide treatment, due to the small number of responsive endodermal cells in total root cells. In planta biochemical validation of MYB36 phosphorylation would require a cell-type-specific enrichment and phosphoproteomics approach. This approach could also help confirm and further delineate the MKK-MPK network coding output specificity in the endodermis.

Methods

Plant materials

Arabidopsis thaliana ecotype Columbia (Col-0) was used as wild-type controls and the background ecotype for all experiments. The mutants fls2 (SALK_062054C)10, sgn3-3 (SALK_043282)17, cif1 cif220(generated by CRISPR-Cas9), myb36-2 (GK- 543B11)19 and pSGN3::SGN3-mVenus in wild type17 were previously described. The 2-in-Endo line was generated in two steps: (1) pSGN3::FLS2-3myc-GFP was introduced into fls2 (SALK_062054C) background and (2) cif1 cif2 null alleles were obtained by CRISPR-Cas9 using a single sgRNA (5′-gctttggtttaggactggag-3′) that targets both genes20,37. The pCASP1::FLS2-3myc-GFP fls2 (SAIL691_C04) described previously32 was crossed with sgn3-3 (SALK_043282) to obtain pCASP1::FLS2-GFP fls2 sgn3.

T-DNA mutant seeds mpk2 (SALK_047422C)94, mpk3 (SALK_151594)95, mpk6 (SALK_073907)95, mpk9 (SALK_064439C), mpk16 (SALK_059737C), mpk17 (SALK_020801C)96, mpk19 (SALK_075D213C), mkk4 (SALK_0188040C)97, mkk5 (SALK_047797C)98 and mkk9 (SAIL_60_H06)52 were provided by the Nottingham Arabidopsis Stock Centre (NASC). Most T-DNA lines in our study were previously analysed and reported to show elimination of transcript/protein levels in the cited publications, with mkk4 (SALK_0188040C) showing reduced transcript level97. All lines were verified for homozygosity. See Supplementary Table 4 for corresponding T-DNA screening primers. Homozygous double mutants mpk2 mpk16 (T-DNA) and mkk4 mkk5 (T-DNA) were generated by crossing the above indicated T-DNA mutants. mpk2 mpk16 double mutant lines #54-9 and #55-13 were generated by CRISPR-Cas9 using MPK2-targeting sgRNA1 (5′-ccgataggccgaggcgcgta-3′) sgRNA2 (5′-tcttttgcatagttgcttcg-3′) and MPK16-targeting sgRNA1 (5′-agagtttcgagacatttacg-3′) sgRNA2 (5′- atacaccgactgcgatattc-3′). mkk4 mkk5 (TILLING)46,99, mkk1 (SALK_027645)100 mkk2 (SAIL 511_H01)101 and mkk3 (SALK_051970)102 were obtained from Jean Colcombet, while mpk3 mpk6 pMPK6::MPK6YG and mpk3 mpk6 pMPK3::MPK3TG were obtained from refs. 45,46.

The published constructs pCASP1::CASP1-GFP with FastRed selection cassette36 and pGPAT5::mCitrine-SYP122 (ref. 53) were introduced into respective backgrounds by floral dipping. For pathway-specific reporters, promoter fusion constructs in Supplementary Table 2 were introduced into the 2-in-Endo background. For the MPK expression atlas (Supplementary Table 3), promoter fusion constructs pMPK1-20::NLS-3xmVenus were introduced into wild-type background.

Plant growth conditions

Plant seeds were surface sterilized in 70% ethanol + 0.05% Tween 20 for 7–10 min, then washed in 96% ethanol and dried in sterile conditions. Seeds were stratified for 2 days at 4 °C in the dark on half-strength Murashige and Skoog (½MS) + 0.8% agar (Roth) plates containing 500 mg l−1 MES buffer (Duchefa). Seedlings were grown vertically for 5–6 days at 23 °C under continuous light before analysis.

Plasmid construction and plant transformation

The In-Fusion Advantage PCR Cloning kit (Clontech) and Multisite Gateway Cloning Technology (Invitrogen) were used for generation of constructs.

All inducible constructs (ind) were assembled by triple Gateway recombination reaction (LR) between entry clones P4-pELTP (LTPG15)-XVE-P1r (promoter)103, L1-HopAI1-L2, L1-MPKa-L2 or L1-MKKa-L2 (gene of interest), and R2-3xFlag-L3 (C-terminal tag) into the destination vector pF7m34GW, containing a FastRed plant selection cassette with a 35S terminator. Complementary (c)DNA entry clones of HopAI1 and HopAI1H103A were synthesized by Genscript41; MPK6a (D218G E222A)104 was obtained from Jean Colcombet; MPK2a (Q188G E192A), MKK1a (T218E S224D), MKK2a (T220D T226E), MKK3a (S235E T241D), MKK4a (T224D S230E), MKK5a (T215E S221E), MKK6a (S221D T227E), MKK7a (S193E S199D), MKK8a (S195D S201E), MKK9a (S195E S201E) and MKK10a (S197E) were cloned from Col-0 cDNA and mutations obtained through site-directed mutagenesis.

All MYB36 constructs were assembled by triple Gateway recombination reaction between entry clones P4-pMYB36(4kb)-R1 (native promoter), L1-MYB36WT-L2, L1-MYB36AAA-L2 or L1-MYB36DDD-L2 (gene of interest), and R2-MYB36-native terminator(250bp)-L3, R2-mCherry-4G-L3 or R2-SuperfolderGFP-TurboID(BirA*)-6His-TEV-3FLAG-L3 (terminator or C-terminal tag) into destination vectors pF7m34GW,0 containing a FastRed plant selection cassette or pFG34GW, containing a FastGreen plant selection cassette. Genomic (g)DNA entry clones of wild-type MYB36 were cloned from Col-0 gDNA, and mutants MYB36AAA (S18A, S146A, S169A) and MYB36DDD (S18D, S146D, S169D) were synthesized by Genscript.

The pathway-specific reporter constructs (Supplementary Table 1) and MPK reporter constructs (Supplementary Table 3) were made, first by generating promoter entry clones using In-Fusion: the endogenous promoter sequences (~2 kb before the start codon) were cloned from Col-0 gDNA or JAtY clones. The reporter constructs were then assembled by double Gateway recombination reaction between a promoter entry clone and L1-nuclear localization signal (NLS) fused to a triple mVenus fluorescent tag (3xmVenus)-L2 or L1-NLS-3xmScarlet-L2 into the destination vector pFG24GW (FastGreen), pFR7m24GW (FastRed) or pB7m24GW (BASTA selection). See Supplementary Tables 4 and 5 for plasmid and primer details.

All plasmids were transformed by heat shock into Agrobacterium tumefaciens GV3101 strain with pSoup plasmid (pMP90) and then transformed into the corresponding plant lines using the floral dip method105,106. On the basis of the plant selection marker, we typically obtained 10–30 transgenic lines for initial characterization or analyses at T1 generation. From these, 2–6 independent T2 transgenic lines were isolated for further characterization. For all T2 analysis in this paper, all plants were pre-selected using fluorescent seedling selection marker. Independent homozygous T3 lines were confirmed by non-segregation of the selection marker and used for western blot experiments or as background for further introduction of constructs. For certain experiments, T1 populations were used to fully capture the inherent expression variability due to random transformation events. This T1 analysis ensures that statistically significant phenotypic differences are indeed reflective of the constructs’ effects themselves and not transgene expression variability. The use of fluorescent seed selection markers (FastRed or FastGreen)107 substantially reduces the occurrence of false positives during our T1 and T2 selection process and facilitates our analysis without the stress of antibiotic selection.

Seedling growth and treatments

The flg22 peptide from Pseudomonas aeruginosa (QRLSTGSRINSAKDDAAGLQIA) was ordered from EZBioLab, and CIF2 peptide (DY*GHSSPKPKLVRPPFKLIPN, * indicates sulfated tyrosine) from Peptide Specialty Laboratories (https://www.peptid.de/). Peptides were dissolved in deionized MilliQ sterile water at the stock concentration of 1 mM and diluted in melted ½ MS agar medium to indicated concentrations. Peptide treatments: seedlings were grown vertically for 4–5 days, then transferred with care onto plates containing peptides or H2O control to grow vertically for 1 day before analysis.

For time-lapse imaging and videos with peptide treatments, 3-day-old seedlings grown vertically on ½ MS plates were transferred to a hydroponic system (a 12-well Multiwell plate (CytoOne) with 6.5 ml liquid ½ MS in each well) to grow for 2 days. These 5-day-old seedlings were then transferred inside a chambered coverglass (Thermo Scientific) and then covered with a ½ MS agar block containing 1 µM peptide. A volume of 50 μl of liquid ½ MS containing the same peptide concentration was added through a channel in the middle of the agar block, directly reaching the roots.

EST (10 mM stock, Sigma, solvent dimethylsulfoxide (DMSO)) and NM-PP1 (Adipogen, solvent DMSO) were diluted to 5 µM in melted ½ MS agar medium, and 0.05% v/v DMSO was used as control treatments. For estradiol (EST) treatments, seedlings were grown vertically on EST or DMSO plates for 5 days or transferred to EST or DMSO to grow for 1 day. For NM-PP1 treatments, seedlings were grown vertically on ½ MS plates for 4 days and transferred to NM-PP1 or DMSO plates to grow vertically for 1 day.

Fluorescence microscopy

Imaging was performed using Leica SP8, Leica Stellaris or Leica Stellaris 5 WLL IR confocal laser scanning microscopes and Leica Thunder light microscope (DM6BZ). Images were taken with a ×63 water immersion objective (Leica SP8, Leica Stellaris) at zoom factor 1 for overviews and zoom factor 5 for surface views. Time-lapse images of Fig. 2f and Supplementary Videos 2 and 3 were taken with a ×63 oil immersion objective (Leica Stellaris 5 WLL IR). Time-lapse images of Extended Data Fig. 3d and Supplementary Video 1 were taken with a ×40 oil immersion objective (Leica Stellaris). Root scans for fluorol yellow (suberin staining) and pGPAT5:mCitrine-SYP122 (suberin synthesis) were acquired using a Leica Thunder light microscope with ×10 dry objective and tile-scan (10% overlap). Unless indicated otherwise, confocal images comparing genotypes/treatments were taken following the ‘four identical criteria’: same position in the roots, the same laser detection intensity, the same laser scanning area (zoom) and the same interval of slices for Z-stack projection. For overview images, maximum projection of z-stacks (step size = 1 µm) are shown. For surface-view images, maximum projection of z-stacks (step size = 0.36 µm) are shown.

The excitation and detection windows were set as follows: for live imaging, GFP (488 nm, 495–550 nm), PI (488 nm, 600–690 nm), mVenus (514 nm, 520–565nm), mCitrine (514 nm, 555–570 nm), mScarlet (561 nm, 580–640 nm) and mCherry (561 nm, 585–680 nm). For time-lapse imaging with Leica Stellaris 5 WLL IR: mVenus (507 nm, 520–560 nm) and mScarlet (555 nm, 580–600 nm). For fixed samples, sequential scanning was used to avoid interference between the following fluorescence channels: calcofluor white (405 nm, 415–435 nm), GFP (488 nm, 495–525 nm) and basic fuchsin (561 nm, 600–650 nm). For suberin analysis on Thunder: fluorol yellow (488 nm, 500–550 nm) and mCitrine (488 nm, 500–550 nm).

Propidium iodide penetration assay

PI penetration assays assess the stage of the formation of a functional CS diffusion barrier15,16. Seedlings were incubated in 10 µg ml−1 PI for 10 min and rinsed in H2O before imaging. PI block was identified as the endodermal cell achieved the exclusion of PI signal from the inner side. The number of endodermal cells was counted from the onset of elongation until PI block. For roots with no functional barrier, PI was not blocked near the root–hypocotyl junction and roots are thus shown as individual data points in the category ‘near hypocotyl’ and excluded from numerical statistical tests.

Suberin quantification assay

Suberin was stained using the methanol-based fluorol yellow protocol20,32. Seedlings were fixed and cleared in methanol for at least 3 days at 4 °C, changing the methanol at least once. The cleared seedlings were transferred to a freshly prepared solution of Fluorol Yellow 088 (0.01% in methanol) and incubated for 1 h at room temperature or overnight at 4 °C in the dark. The stained seedlings were rinsed in methanol and transferred to a freshly prepared solution of aniline blue (0.5% in methanol) for 1 h counterstaining in the dark. Finally, the seedlings were washed briefly in water before mounting and imaging with the Leica Thunder light microscope.

Fixation and lignin staining

Fixation and staining were performed using an adapted Clearsee protocol108,109. Briefly, seedlings were fixed in 4% paraformaldehyde PBS solution at 4 °C overnight using 6-well or 12-well plates (CytoOne), then washed twice for 1 min with PBS. Once fixed, seedlings were cleared in Clearsee solution for at least 24 h under gentle shaking. Fixed and cleared samples were then incubated overnight in a Clearsee solution supplemented with 0.2% basic fuchsin and 0.1% calcofluor white for combined cell wall and lignin staining. Once the dye solution was removed, samples were rinsed overnight in Clearsee twice before mounting and observation using confocal microscopes.

Electron microscopy for ROS and lignin detection

Visualization of H2O2 (ROS) around the CS was performed with the cerium chloride assay described previously20,37. This detection method is based on Ce3+ ions reacting with H2O2, forming electron-dense cerium perhydroxide precipitates, which are detected by transmission electron microscopy. Briefly, seedlings were incubated in 50 mM MOPS buffer (pH 7.2) containing freshly prepared 10 mM cerium chloride (CeCl3) (Sigma) for 45 min. Seedlings were then washed twice in MOPS buffer for 5 min and fixed in 2.5% glutaraldehyde solution (EMS) in 100 mM phosphate buffer (pH 7.4) for 1 h at room temperature. Then, they were postfixed in 1% osmium tetroxide (EMS) with 1.5% potassium ferrocyanide (Sigma) in phosphate buffer for 1 h at room temperature. The plants were rinsed twice in distilled water and then dehydrated in ethanol solution (Sigma) at gradient concentrations (30%, 40 min; 50%, 40 min; 70%, 40 min; 100%, 1 h ×2). This was followed by infiltration in Spurr resin (EMS) at gradient concentrations (Spurr 33% in ethanol, 4 h; Spurr 66% in ethanol, 4 h; Spurr 100%, 8 h ×2) and finally polymerized for 48 h at 60 °C in an oven. Ultrathin sections (50 nm) were cut transversally at 1.5, 2 or 3 mm from the root tip on a Leica Ultracut ultramicrotome (Leica Mikrosysteme) and picked up on a 2 × 1 mm nickel slot grid (EMS) coated with a polystyrene film (Sigma).

Visualization of lignin deposition around the CS and cell corner was done using the permanganate potassium (KMnO4, Sigma) staining protocol110,111. The sections were post-stained with 1% KMnO4 in H2O for 45 min and rinsed several times with H2O.

Micrographs were taken with a CM100 transmission electron microscope (FEI) at an 80 kV acceleration voltage with a TemCamF416 digital camera (TVIPS) using the software EM-MENU 4.0 (TVIPS). Panoramic views were aligned using the software IMOD112.

Ectopic ROS quantification from EM images

For Extended Data Fig. 7d EM ROS quantification, all samples from the selected lines—indMKK2a, indMKK4a, indMPK6a and indMPK2a in sgn3 CASP1-GFP with and without estradiol induction were collected, prepared and imaged in a single experiment. We assessed 5 roots per sample and quantified the ROS staining patterns categorically cross an average of 42 endodermal cells per sample: from the TEM images, each endodermal cell was carefully examined for ROS staining patterns (electron-dense dots and patches) occurring outside of the Casparian strip region, typically between the endodermis–cortex boundary. These ectopic ROS patterns were categorized as no, yes low or yes strong on the basis of their intensity.

RNA-seq experiments and analysis

Sample preparation for RNA-seq experiments was as previously described20. Briefly, wild-type, sgn3, fls2 and pCASP1::FLS2-GFP fls2 seeds were grown vertically on a sterile mesh on solid ½ MS plates for 5 days and transferred onto fresh ½ MS solid medium containing 1 μM peptides and mock plates (mock and peptide treatments always transferred in parallel). After 30-, 120- and 480-min incubation on plates, whole roots from 2 square plates (50 ml) were cut and collected for each sample, then immediately frozen in liquid nitrogen. Three biological replicates for each sample were conducted on three different days. Total RNA was extracted with a TRIzol-adapted ReliaPrep RNA extraction kit (Promega).

Libraries were prepared by the Lausanne Genome Technology Facility following the same protocol20. RNA quality control was performed on a Fragment Analyzer (Advanced Analytical Technologies). Total RNA (1,000 ng) was used to prepare RNA-seq libraries with the Illumina TruSeq Stranded mRNA reagents (Illumina) on a Sciclone liquid handling robot (PerkinElmer) using a PerkinElmer-developed automated script. The resulting library was used for cluster generation with the Illumina TruSeq SR Cluster Kit v4 reagents and sequenced on the Illumina HiSeq 2500 system using TruSeq SBS Kit v4 reagents. The Illumina Pipeline Software v.2.20 was used to process sequencing data.

The Lausanne Genomic Technologies Facility performed the data processing using their in-house RNA-seq pipeline, as described in ref. 20. Briefly, purity-filtered read trimming for adapters and low-quality sequences was done with Cutadapt (v.1.8)113 and removal of reads matching ribosomal RNA sequences with fastq_screen (v.0.11.1). Low complexity reads were filtered with reaper (v.15-065)114. Cleaned reads were aligned against A. thaliana TAIR10 genome using STAR (v.2.5.3a)115 and read counts per gene locus were obtained with htseqcount (v.0.9.1)116 using A. thaliana TAIR10 Ensembl 39 gene annotation. RSeQC (v.2.3.7)117 was used to evaluate the quality of the data alignment.

Statistical analysis for genes was performed in R (3.5.3). Genes with low counts were filtered out according to the rule of one count per million (cpm) in at least one sample. Library sizes were scaled using trimmed mean of M values normalization and log transformed into cpm (EdgeR package v.3.24.3)118. Data were corrected for the experimental batch effect using the removeBatchEffect function (limma). Statistical quality controls were performed through pairwise sample correlations, clustering and sample Principal component analysis using batch corrected normalized data. Differential expression was computed using the limma-trend approach by fitting all samples into one linear model119. The experimental batch factor was added to the model. Two-sided moderated t-test was used for each pairwise comparison (peptide-treated vs H2O-treated) per timepoint. The adjusted P value was computed using the Benjamini–Hochberg method, controlling for false discovery rate. To determine genes significantly regulated by CIF2 or flg22 (*s in Fig. 2 and Extended Data Fig. 2), the adjusted P value was set to be below 0.05 (Padj < 0.05).

Heat maps of gene lists in Fig. 2c,d were selected on the basis of published genes involved in lignin and suberin biosynthesis20,120. Figure 2e genes were based on the intersection of RNA-seq datasets in ref. 30 and microarray datasets in ref. 19 that report significant downregulation in myb36 mutants compared with wild type. myb36 vs wild type regulation heat map (left) was generated using the data in ref. 30. Figure 2b genes were based on genes significantly upregulated by flg22 in whole seedlings41. The heat map of wild type treated with flg22 (left) in Extended Data Fig. 2b was generated using the data in ref. 41.

GO-enrichment analyses were performed using the clusterProfiler R package (3.18.1) with ‘Biological process’, and P value was adjusted using the Benjamini–Hochberg method with a cut-off of 0.05. GO annotations used the org.At.tair.db R package (3.12.0). Gene ratio represents the gene counts in a GO category that are differentially regulated by a pathway divided by the total number of genes in that GO category.

For Fig. 4a and Supplementary Fig. 7, seedlings were grown vertically for 5 days on sterilized mesh on solid ½ MS media. The seedlings were treated by adding 20 ml liquid ½ MS containing respective peptides or H2O control to submerge the roots. At 5, 10 and 15 min, liquid was drained and roots were cut (Fig. 4a), or whole seedlings were collected (Source Data Supplementary Fig. 7b). For Extended Data Fig. 5b and 6d, seedlings were grown for 5–6 days on sterilized mesh on solid ½ MS media supplemented with EST or DMSO.

Root tissues collected were immediately frozen in liquid nitrogen and ground using Tissue Lyser II (Qiagen). Extraction buffer (50 mM Tris HCl pH 7.5, 150 mM NaCl, 0.5 mM EDTA, 1% Triton X-100, 50 mM β-glycerophosphate, 1 mM phenylmethylsulfonyl fluoride, 100 μM Na orthovanadate, 5 mM Na fluoride, 2.5 mM MgCl2, 1x complete EDTA-free protease inhibitor cocktail) was added to the frozen samples. The samples were briefly mixed by vortexing and centrifuged at 21,000 × g for 15 min at 4 °C. The supernatant was transferred to new tubes and the total protein concentration was measured using the Bradford method (Thermo Fisher). After adjusting total protein concentration, the proteins were denatured by addition of 3x (v/w) NuPAGE LDS sample buffer + 10 mM dithiothreitol (Invitrogen, NP0007) and heated for 5 min at 95 °C.

Equal amounts of protein were loaded (~20–40 μg per lane) and separated on 10% or gradient (4–12%) pre-cast acrylamide gels (Eurogentec). After electrophoresis, the separated proteins were transferred onto PVDF membranes (Immobilon-E, Merck-Millipore) using Pierce FastBlotter G2 semi-dry blotting (Thermo Fisher). The blot was blocked in 1% blocking buffer in TBS (western blocking reagent, Roche) for 1 h at room temperature before incubation with primary antibodies: anti-p44/42 (1:1,000, Cell Signaling Technologies, 4370), anti-MPK6 (1:10,000, Sigma, A7104), anti-FLAG conjugated M2 peroxidase (1:500, Sigma, A8592) and anti-actin (1:2,000, Sigma, A0605) in 0.5% blocking buffer + TBS-0.05% Tween overnight at 4 °C. After washing the blot two to three times with 1x TBS + 0.1% Tween (TBS-T), the blot was treated with secondary antibodies (anti-rabbit, 1:10,000, Agrisera for anti-p44/42; anti-rabbit HRP ECL, 1:30,000 for anti-MPK6; anti-mouse HRP, 1:5,000 for actin, Promega) in 0.5% blocking buffer + TBS-0.05% Tween for 2 h and then washed 5–7 times with TBS-T. For loading control detection of MPK6 and actin, the original membrane was stripped (Thermo Scientific). Signal was detected with SuperSignal West Pico Plus and Femto kits (Thermo Scientific).

Quantification and statistical analysis

All fluorescence microscopy images were processed using Fiji (ImageJ2 v.2.9.0/1.53t) with Fiji macros to semi-automate the processing and quantification121. Macros will be made available on request.

Percentages (%) of gaps in CASP1-GFP (Fig. 1e and Extended Data Fig. 1h) were based on maximum projections of surface-view images (GFP channel) after thresholding to distinguish background from signal. Quantification represents the percentage of uncovered area (background) of a fused CASP1-GFP belt reconstructed from connecting individual domains using a selection Bush tool. Fully connected CASP1-GFP yields 0% of gaps.

Numbers of holes per 100 µm of CASP1-GFP (Figs. 3e, 4e, 5c, 6e and Extended Data Fig. 1c) were based on maximum projections of overviews (GFP channel) after thresholding to distinguish background from signal. Steps for quantification are detailed in Extended Data Fig. 1b. Each data point represents the average number of holes per 100 µm calculated from a single overview image.

Quantifications of total grey intensity of nuclei signals (Fig. 2f) were calculated from ‘sum slice projection’ of overview images at each hour. This method sums the intensity values at each pixel location throughout the stack and projects the sum onto a two-dimensional image. This is useful for visualizing the total intensity contribution from all slices in the stack, especially in the cases of overlapping nuclei. Thresholding and region of interest selection were then applied to isolate nuclei signals from noise and background. Similar procedures were used for Fig. 6d to calculate total grey intensity of MYB36 nuclei signals from maximum projection of overview images.

For quantification of CASP1 domain properties: number of fragments per 100 µm (Fig. 3c) and the average size of fragments (Extended Data Fig. 3c) were based on the same data points from maximum projections of surface-view images. Thresholding followed by particle analysis (no minimum size) was used to quantify the number of fragments and the average size of fragments in a single image. The length of CASP1 in µm for a single image was measured and used to calculate the number of fragments per 100 µm.

Quantification of CS coverage (Figs. 5d, 6b and Extended Data Fig. 6e) is represented by the percentage of CASP1 length coverage normalized to wild type. Total CASP1 length (measured after applying skeletonize (Fiji)) of each overview image (maximum projection) was measured and then normalized to the average CASP1-GFP length in wild-type roots from the same experiments to calculate the percentage of CS coverage for each data point.

All graphs and statistical analyses were performed using R 4.0.4 (http://www.R-project.org/). For multiple comparisons within the same dataset, analysis of variance (ANOVA) followed by Tukey’s honestly significant difference (HSD) test was applied when linear model assumptions were met. Unbalanced Tukey’s test was used for unequal replication. For comparing the mean of one sample with a control or treatment versus no treatment, Welch’s t-test was used: for multiple comparisons using t-tests, P values were adjusted by Bonferroni correction. In boxplots, boxes span the first to third quartiles, with the bold line representing the median. Whiskers indicate maximum and minimum values, except outliers (defined as >1.5× interquartile range above the third quartile or below the first quartile, respectively). Outliers are always shown as dots. Bold lines in dot plots represent the mean. Each point in the boxplot or dot plot represents a data point from an individual root (n). Statistical parameters, error bars and n are reported in the Figure legends.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.