banner
Home / News / Extracellular plant subtilases dampen cold-shock peptide elicitor levels | Nature Plants
News

Extracellular plant subtilases dampen cold-shock peptide elicitor levels | Nature Plants

Oct 16, 2024Oct 16, 2024

Nature Plants (2024)Cite this article

186 Accesses

Metrics details

Recognizing pathogen-associated molecular patterns on the cell surface is crucial for plant immunity. The proteinaceous nature of many of these patterns suggests that secreted proteases play important roles in their formation and stability. Here we demonstrate that the apoplastic subtilase SBT5.2a inactivates the immunogenicity of cold-shock proteins (CSPs) of the bacterial plant pathogen Pseudomonas syringae by cleaving within the immunogenic csp22 epitope. Consequently, mutant plants lacking SBT5.2a activity retain higher levels of csp22, leading to enhanced immune responses and reduced pathogen growth. SBT5.2 sensitivity is influenced by sequence variation surrounding the cleavage site and probably extends to CSPs from other bacterial species. These findings suggest that variations in csp22 stability among bacterial pathogens are a crucial factor in plant–bacteria interactions and that pathogens exploit plant proteases to avoid pattern recognition.

Pathogen recognition is pivotal to plant survival. Pathogen-associated molecular patterns (PAMPs) are recognized on the plant cell surface by pattern-recognition receptors (PRRs). Upon PAMP binding, PRRs associate with receptor kinases and intracellular protein kinases to activate pattern-triggered immunity (PTI), which includes the production of reactive oxygen species (ROS) and ethylene, apoplast alkalinization, MAP kinase phosphorylation and transcriptional reprogramming1,2,3.

PAMP recognition by PRRs occurs in the extracellular space within plant tissues (the apoplast). The apoplast is the first and often final destination for pathogens and is an important site for pathogen proliferation3. Pathogen colonization of the apoplast is partly mitigated by plant-secreted hydrolases, including proteases, which are secreted both constitutively and inducibly4,5. Ser proteases, including subtilases, are the largest class of secreted proteases4.

PAMPs can be oligosaccharides, lipids and peptides, and a wide range of peptide-based PAMPs have been identified from fungal, oomycete and bacterial pathogens6. PRRs tend to perceive a conserved epitope of a PAMP that has important functions to the microorganism7. Although the release of immunogenic PAMP peptides from their precursors could be an essential step in pathogen recognition, our knowledge of the biogenesis and maintenance of PAMPs and the involved enzymes is very limited8.

The conserved nucleic acid binding motif RNP-1 of bacterial cold-shock proteins (CSPs) serves as a PAMP that is perceived by PRRs from several Solanaceae species, including tomato (Solanum lycopersicum), tobacco (Nicotiana tabacum) and potato (S. tuberosum)9,10,11. CSPs are highly induced in bacteria in response to rapid downshifts in temperature, which is the basis for their name. However, CSPs are also activated by other types of stress, and many are constitutively produced12. The 22-residue amino-terminal consensus of 150 bacterial CSP sequences from genera such as Micrococcus, Bacillus and Escherichia was introduced as csp22. Residues 5–19 of csp22 (csp15) constitute the active PAMP epitope that can trigger plant immune responses, including a burst of ROS9,10. Two distinct receptors, the receptor-like protein CSPR and the receptor-like kinase CORE, were claimed to act as CSP receptors in older Nicotiana benthamiana plants10,11, but the affinity of csp22 for CORE is much higher than that for CSPR11, and only the silencing of CORE makes these plants insensitive to csp22 (ref. 13). CSPR was later identified as RE02 and is involved in perceiving small Cys-rich proteins from Valsa mali14 and Sclerotinia sclerotiorum15.

In this study, we tested the hypothesis that extracellular plant proteases might play important roles in the biogenesis of the CSP elicitor of the model bacterial pathogen Pseudomonas syringae pv. tomato DC3000 (PtoDC3000). Surprisingly, however, we discovered proteolytic degradation of csp22 in the apoplast, mediated by serine protease SBT5.2, which dampens the CSP-triggered immune response and suggests that pathogens take advantage of plant proteases to escape PAMP recognition.

To determine the role of CSP elicitor release in bacterial immunity, we studied the release of csp22 from PtoDC3000. The PtoDC3000 genome encodes six CSPs that contain the cold-shock domain (PF00313) and comprehend peptides similar to the csp22 consensus (Fig. 1a). Five of these six CSP-encoding genes are expressed during infection16 (Fig. 1b). Since the original discovery of csp22 included the CspD and CapB proteins from various bacteria9, we expressed these two proteins in Escherichia coli with a His tag linked via the TEV cleavage sequence to the N terminus. Heterologous expression only delivered sufficient CspD protein (Extended Data Fig. 1), so we continued our studies with CspD.

a, Sequence alignment of the six CSPs of PtoDC3000 with the frequently used consensus csp22 peptide. b, Transcript levels of CSPs of PtoDC3000 during infection in counts per million (CPM). Extracted from ref. 16. c, CspD-derived csp22 triggers an oxidative burst in six-week-old N. benthamiana plants. d, CspD-derived csp22 triggers only a weak oxidative burst in four-week-old N. benthamiana plants. e, The oxidative burst triggered by csp22 of CspD in six-week-old plants is absent when NbCORE is silenced. f, The weak oxidative burst triggered by csp22 of CspD in four-week-old plants is absent when NbCORE is silenced. In c–f, one-week-old N. benthamiana plants were infected with and without TRV carrying a fragment of GUS (TRV::GUS) or NbCORE (TRV::CORE). Leaf discs of four- and six-week-old plants were treated with water or 500 nM csp22, and ROS were measured in relative light units (RLU). The lines represent the mean and the shading represents the s.e.m. of n = 12 (c,d) or n = 6 (e,f) leaf discs.

The csp22 peptide of CspD differs at nine residues from the consensus csp22 reported originally9. To test whether the csp22 peptide of CspD is recognized by N. benthamiana, we synthesized this 22-residue peptide and added it to leaf discs of six-week-old N. benthamiana floating on a solution containing luminol and horseradish peroxidase (HRP). Luminescence measurements revealed that csp22 of CspD is able to trigger a classic oxidative burst by releasing ROS, in contrast to the water control (Fig. 1c). Only a weak oxidative burst was detected in four-week-old plants (Fig. 1d), consistent with the low expression of NbCORE in younger plants11.

To confirm that NbCORE is required for detecting csp22 of CspD, we depleted NbCORE with virus-induced gene silencing (VIGS) using tobacco rattle virus (TRV) vectors carrying a 300-bp fragment of NbCORE, or of β-glucuronidase (GUS) as a negative control13. Oxidative burst assays showed that six-week-old TRV::CORE plants are blind to csp22 of CspD, in contrast to TRV::GUS plants, which show a csp22-induced oxidative burst (Fig. 1e). The weak csp22-induced response detected in four-week-old TRV::GUS plants is also absent from TRV::CORE plants (Fig. 1f), indicating that weak responses in younger plants are still NbCORE-dependent.

We next tested whether the purified CspD protein (Fig. 2a) could also trigger an NbCORE-dependent oxidative burst. We expressed CspD with an N-terminal His tag in E. coli and purified this over Ni-NTA (Fig. 2a). Indeed, CspD used at similar concentrations as csp22 triggers an oxidative burst in six-week-old TRV::GUS plants, but not in TRV::CORE plants (Fig. 2b). The oxidative burst was nearly absent in four-week-old TRV::CORE and TRV::GUS plants (Fig. 2c), consistent with the low NbCORE expression in younger plants. These data demonstrate that purified CspD triggers an NbCORE-dependent oxidative burst, indicating that this sample does not contain other elicitors such as the bacterial flagellin of E. coli, which could also have triggered an oxidative burst in leaf discs of N. benthamiana because they express NbFLS2.

a, Purification of CspD from P. syringae heterologously expressed in E. coli. PtoCspD was tagged with an N-terminal His–TEV tag, expressed in E. coli and purified by immobilized metal affinity chromatography. This experiment was repeated two times with similar results. b, CspD triggers an oxidative burst in six-week-old plants that is absent when NbCORE is silenced. c, CspD does not trigger an oxidative burst in four-week-old plants. In b,c, one-week-old N. benthamiana plants were infected with TRV carrying a fragment of GUS (TRV::GUS) or NbCORE (TRV::CORE). Leaf discs of four- and six-week-old plants were treated with 500 nM CspD, and ROS were measured in RLU. The lines represent the mean and the shading represents the s.e.m. of n = 6 leaf discs.

To test whether apoplastic fluids (AFs) could process CspD, we incubated purified CspD with AF isolated from four- to six-week-old N. benthamiana leaves and analysed proteins by SDS–PAGE. This experiment revealed a quick disappearance of intact CspD protein within 15 minutes (Fig. 3a), indicating that CspD is unstable in AF.

a, Purified CspD is quickly degraded in AF of N. benthamiana. Purified CspD (2 μM) was incubated with water or AF for various times, and the products were analysed on a protein gel via Coomassie staining. This experiment was repeated four times, with similar results. b, Degradation products of CspD in AF detected by LC–MS/MS analysis. Purified CspD was incubated with AF isolated from N. benthamiana for 15 and 60 minutes. Proteins were precipitated with 80% acetone, and the peptide fraction (supernatant) was dried and analysed via LC–MS/MS. All CspD-derived peptides detected after 15 minutes of incubation were aligned with the CspD protein sequence. The red shaded region indicates csp22 of CspD. Likely initial cleavage sites are indicated by S1 and S2. The bottom graph shows the coverage of the CspD protein after incubation for 15 and 60 (dotted line) minutes, generated by counting the number of times that each residue was detected in the peptides. Peptide coverage graphs of 15 and 60 minutes’ incubation and the water controls are shown in Extended Data Fig. 2. c, Proposed degradation of CspD, first by endopeptidase(s) at sites S1 and S2 and then by exopeptidases. Cleavage at S1 inactivates the csp22 elicitor peptide. d, Positions of the S1 and S2 sites in the structural model of CspD. This structural model was obtained from the AlphaFold-predicted structure of CspD in the Uniprot database (entry Q87ZR9), trimmed for the cold-shock domain, with the csp22 peptide (orange) and S1 and S2 cleavage sites (red) as indicated.

Source data

To further investigate CspD degradation in AF, we analysed the released peptides via liquid chromatography–tandem mass spectrometry (LC–MS/MS) and mapped the CspD-derived peptides onto the CspD protein sequence. A total of 171 different CspD-derived peptides were detected, covering the entire CspD protein sequence (Fig. 3b). The peptides overlap and are staggered in three clusters, differing only in single residues removed from the N and carboxy termini (Fig. 3b and Extended Data Fig. 2). This peptide pattern indicates that CspD is cleaved at two sites (S1 and S2) by endopeptidases cleaving at VKWF ↓ NNAK and YKTL ↓ KAGQ, and that exopeptidases subsequently remove a series of single residues from either end (Fig. 3c). Cleavage at site S1 would cleave 10.0 kDa CspD into fragments of 1.3 and 8.7 kDa, whereas processing at S2 would result in 5.1 and 4.9 kDa fragments (Fig. 3c). Analysis of the CspD structural model8 indicates that site S1 is in the exposed β-sheet in middle of the csp22 elicitor sequence, and site S2 is in a nearby exposed loop (Fig. 3d).

Cleavage at site S1 of CSP within the csp22 epitope was unexpected and would inactivate the immunogenicity of csp22. To confirm csp22 cleavage, we incubated the csp22 peptide briefly in AF and then added this to leaf discs of csp22-responsive plants to detect remaining elicitor activity. Remarkably, elicitor activity drastically diminished within five minutes and was absent after 15 minutes of incubation (Fig. 4a), demonstrating that csp22 is unstable in AF.

a, Incubation in AF quickly inactivates the csp22 elicitor. AF was incubated with 500 nM csp22 peptide for 5 or 15 minutes and then added to leaf discs of six-week-old N. benthamiana plants floating on the luminol–HRP solution. Luminescence (RLU) was measured for 60 minutes with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 12 replicates. b, Conceptual diagram of Qcsp8 showing the cleavage in csp22. c, Qcsp8 is quickly cleaved in AF isolated from N. benthamiana leaves. Qcsp8 (10 μM) was incubated in water or AF, and fluorescence was measured over time. The shading represents the s.e.m. of n = 3 replicates. RFU, relative fluorescence units.

To confirm cleavage in the middle of csp22, we obtained a custom-synthesized quenched octapeptide containing the eight residues surrounding the S1 site (VKWF ↓ NNAK) (Qcsp8). Cleavage of Qcsp8 would separate the N-terminal quencher (DABCYL) from the C-terminal fluorophore (EDANS), causing fluorescence (Fig. 4b). Incubation of Qcsp8 in AF causes a rapid increase of fluorescence compared with the water control (Fig. 4c), indicating that this octapeptide is cleaved in AF.

Being the largest class of proteases detected in the apoplast of N. benthamiana17, subtilases were tested for degrading csp22/CspD/Qcsp8 by taking advantage of subtilase inhibitor Epi1 of the oomycete potato blight pathogen Phytophthora infestans18. Elicitor activity was detected after incubation of csp22 peptide with AFs isolated from leaves transiently expressing Epi1 (AF(Epi1)), but not from leaves transiently transformed with the empty-vector control (AF(EV)) (Fig. 5a). This indicates that Epi1 blocks AF degradation of csp22, implying that apoplastic subtilases are responsible for degrading csp22.

a, Inactivation of csp22 is suppressed in AF of plants overexpressing subtilase inhibitor Epi1. b, CspD degradation is reduced in AF of plants overexpressing subtilase inhibitor Epi1. This experiment was repeated once, with similar results. c, Cleavage of Qcsp8 is reduced in AF of plants overexpressing subtilase inhibitor Epi1. d, Inactivation of csp22 is suppressed in AF isolated from TRV::SBT5.2 plants. e, CspD degradation is reduced in AF isolated from TRV::SBT5.2 plants. f, Cleavage of Qcsp8 is reduced in AF isolated from TRV::SBT5.2 plants. This experiment was repeated two times, with similar results. In a,d, AF was incubated with 500 nM csp22 peptide for 60 minutes and then added to leaf discs of six-week-old N. benthamiana plants floating on a luminol–HRP solution. Luminescence (RLU) was measured for 60 minutes with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 6 (a) and n = 8 (d) replicates. In b,e, AF was incubated with 2 μM CspD protein for various time periods, separated on a 15% SDS–PAGE gel and stained with Coomassie. In c,f, AF was incubated with 10 µM Qcsp8 while fluorescence was measured with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 4 samples.

Source data

We also tested whether CspD protein is more stable in the presence of Epi1 and found that CspD protein is stabilized in AF containing Epi1 and still degraded in AF of the EV control (Fig. 5b). Likewise, AF containing Epi1 was no longer able to cleave Qcsp8, unlike the AF(EV) control (Fig. 5c), indicating that subtilases are responsible for Qcsp8 cleavage. Taken together, these data indicate that subtilases are responsible for inactivating csp22, degrading CspD protein and cleaving Qcsp8.

As the most active subtilase in the apoplast of N. benthamiana17,18,19,20,21,22,23, subtilase SBT5.2 was further investigated for its role in csp22 degradation. We used VIGS to suppress NbSBT5.2 expression in N. benthamiana21,23 and extracted AF from TRV::SBT5.2 plants. N. benthamiana expresses three SBT5.2 homologues (a–c), which are all targeted with the 300-bp fragment of SBT5.2a present in TRV::SBT5.2 (ref. 23). Elicitor activity of csp22 was detected upon incubation in AF from TRV::SBT5.2 plants, but not in AF of TRV::GUS control plants (Fig. 5d), indicating that SBT5.2 is required for inactivating csp22. Likewise, CspD protein was more stable in AF of TRV::SBT5.2 plants than in AF of TRV::GUS control plants (Fig. 5e), and the Qcsp8 peptide was no longer cleaved in AF of TRV::SBT5.2 plants, unlike in the TRV::GUS control (Fig. 5f). Taken together, these data indicate that apoplastic subtilase SBT5.2 is required for inactivating csp22, degrading CspD protein and cleaving Qcsp8.

To determine whether SBT5.2 is also sufficient for inactivating csp22, degrading CspD protein and cleaving Qcsp8, we cloned the open reading frame encoding SBT5.2a (NbD038072 in the NbDE database24; NbL13g04590 in the LAB360 database25) fused to a C-terminal His tag into a binary vector and purified this protein from the AF of agroinfiltrated plants on Ni-NTA columns (Extended Data Fig. 3), resulting in purified SBT5.2a–His protein (Fig. 6a). We detected multiple isoforms that are labelled with the activity-based Ser hydrolase probe FP-TAMRA (Fig. 6a), indicating that they are derived from active proteases. Incubation of csp22 with different concentrations of purified SBT5.2a–His resulted in the dose-dependent inactivation of csp22 elicitor activity (Fig. 6b). Likewise, incubation of CspD with different concentrations of purified SBT5.2a–His resulted in the dose-dependent cleavage of CspD into a smaller isoform that was probably caused by cleavage at the S1 site because that cleavage would result in an 8.7 kDa product, whereas processing at S2 would generate two ~5 kDa products (Fig. 6b). Finally, incubation of Qcsp8 with different concentrations of purified SBT5.2a–His resulted in the dose-dependent cleavage of Qcsp8 (Fig. 6d). By contrast, Qcsp8 was not cleaved by purified P69B–His (Fig. 6e), a subtilase from tomato21, demonstrating the specificity of Qcsp8 cleavage by SBT5.2a. Taken together, these data indicate that apoplastic subtilase SBT5.2a can inactivate csp22, process CspD protein and cleave Qcsp8.

a, Purified SBT5.2a–His. His-tagged SBT5.2a was transiently expressed in N. benthamiana by agroinfiltration and isolated from AF at day 6 on Ni-NTA columns. Proteins were labelled with and without 0.2 μM FP-TAMRA for one hour and separated on SDS–PAGE stained with Coomassie or scanned for in-gel fluorescence. This experiment was repeated four times, with similar results. b, Purified SBT5.2a–His inactivates the csp22 elicitor. Purified SBT5.2a–His was incubated with 100 nM csp22 peptide for 45 minutes and then added to leaf discs of six-week-old N. benthamiana plants floating on a luminol–HRP solution. Luminescence (RLU) was measured for 60 minutes with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 6 replicates. c, Purified SBT5.2a–His processes purified CspD protein. Purified CspD protein was incubated with various concentrations of purified SBT5.2a–His at room temperature for 40 minutes, separated on SDS–PAGE and stained with Coomassie. This experiment was repeated three times, with similar results. d, SBT5.2a cleaves Qcsp8. Various concentrations of purified SBT5.2a–His were incubated with 10 μM Qcsp8 while fluorescence was measured using a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 3 replicates. e, Qcsp8 is cleaved by SBT5.2a–His but not by P69B–His. 0.5 μg of purified SBT5.2a–His and P69B–His were incubated with 10 μM Qcsp8 while fluorescence was measured using a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 4 replicates.

Source data

To investigate the role of SBT5.2 subtilases in immunity, we took advantage of our recently introduced triple-knockout line lacking all three SBT5.2 genes generated by genome editing23. AFs from these mutants are unable to cleave Qcsp8 (Fig. 7a) or inactivate csp22 (Fig. 7b). To assess the role of SBT5.2 in csp22-mediated immunity, we used the fliC mutant of PtoDC3000ΔhopQ (PtoDC3000ΔhopQΔfliC26) to avoid responses triggered by flagellin/FLS2 signalling. Infection assays, however, did not reveal altered bacterial growth in the sbt5.2 mutant lines compared with wild-type (WT) plants (Fig. 7c). Elicitation with csp22 also did not reveal any changes in the oxidative burst (Extended Data Fig. 4a), even at low csp22 concentrations (Extended Data Fig. 4b). We then tested whether immune priming27 by csp22 is enhanced in sbt5.2 mutants. Pretreatment with 1 µM csp22 followed by pathogen inoculation after 24 hours reduced bacterial growth in both sbt5.2 mutant lines compared with WT plants (Fig. 7c). Likewise, pretreatment with 5 μM csp22 also reduced bacterial growth of the fliC mutant of Pta6605 (Pta6605ΔfliC28) more in sbt5.2 mutant plants than in WT plants (Extended Data Fig. 5). These data demonstrate that the degradation of csp22 by SBT5.2 in WT plants increases plant susceptibility to bacterial infection.

a, Qcsp8 is no longer cleaved in AF of sbt5.2 mutants. Qcsp8 (10 µM) was incubated with AF of WT and sbt5.2 mutant plants, and fluorescence was measured with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 4 samples. b, Csp22 inactivation is reduced in AF isolated from both sbt5.2 triple mutants. AFs of WT or sbt5.2 mutant plants were incubated with 500 nM csp22 peptide for 30 minutes and then added to leaf discs of six-week-old N. benthamiana plants floating on luminol–HRP solution. Luminescence (RLU) was measured for 60 minutes with a plate reader. The lines represent the mean and the shading represents the s.e.m. of n = 12 replicates. c, Immunity induced by low csp22 concentration is increased in the sbt5.2 mutant. Leaves of WT and sbt5.2 mutant N. benthamiana were infiltrated with water or 1 μM csp22 and incubated for 24 hours and then injected with 1 × 105 bacteria per ml of the flagellin mutant strain of P. syringae pv. tomato DC3000 (PtoDC3000ΔfliCΔhopQ). Colony-forming units (CFU) were determined one day post infection. The error bars represent the s.e.m. of n = 6 replicates. The P values are from an unpaired, two-tailed Student’s t-test.

To investigate whether other csp22 elicitor peptides present in CSPs of PtoDC3000 have differential sensitivity to SBT5.2-mediated inactivation, we tested two more csp22 peptides that are distinct from csp22 of CspD: csp22 of CapB (PSPTO4145) and PSPTO3984, which differ by 11 and 10 residues from csp22 of CspD, respectively (Fig. 8a). Both csp22 peptides trigger ROS bursts in leaf discs of six-week-old N. benthamiana plants (Extended Data Fig. 6). All three peptides are inactivated when incubated in AF, but the speed of inactivation differs per peptide (Fig. 8b). When incubated with AF of WT plants, the csp22 peptide of PSPTO3984 is more quickly degraded than that of CspD, whereas the degradation of csp22 of CapB is slower than that of CspD (Fig. 8b). However, all peptides are stabilized to similar levels when incubated in AF of the sbt5.2 mutant (Fig. 8b), indicating that SBT5.2 subtilases contribute to the degradation of all three csp22 peptides. Because the residues preceding the cleavage site are identical between CspD, PSPTO3984 and CapB, the reduced processing of CapB by SBT5.2 is probably caused by the presence of acidic residues (Asp16 and Glu17) after the cleavage site. These data indicate that the polymorphism in csp22 peptides can lead to varying sensitivity to SBT5.2-mediated cleavage, resulting in different stabilities in the plant apoplast. The alignment with csp22 peptides from different bacterial plant pathogens indicates that the variation in csp22 stabilities might be similar between different bacterial plant pathogens (Fig. 8c).

a, Alignment of csp22 sequences from CspD (PSPTO03355), PSPTO3984 and PSPTO4145 from PtoDC3000. b, The csp22 peptides (1,000 nM) were incubated with AF from WT plants and the sbt5.2#4 mutant for 5, 60 and 120 min, diluted tenfold and then added to leaf discs of N. benthamiana floating in HRP and luminol. ROS burst was measured immediately with a plate reader over 45 minutes. The lines represent the mean and the shading represents the s.e.m. of n = 8 replicates. c, Phylogeny of CSPs with an alignment of enclosed csp22 sequences of PtoDC3000 (red), P. syringae pv. syringae B728a (PsyB728a; green) and P. syringae pv. phaseolicola 1448A (Pph1448a; orange), as well as Xanthomonas campestris ATCC33913 (cyan) and Ralstonia solanacearum GMI1000 (blue). The highlighted residues are identical (dark grey) or similar (light grey) to the csp22 sequence of CspD. In a,c, arrowhead indicates putative cleavage site.

This work has demonstrated that immunogenic csp22 peptides are quickly inactivated by subtilase SBT5.2 in the apoplast. This discovery indicates that SBT5.2 dampens PTI by degrading csp22 and that bacteria may take advantage of SBT5.2 degrading elicitors to evade recognition.

This project was initiated to investigate whether peptide elicitors require proteolytic processing for their release from their precursors. We anticipated this since most peptide elicitors are not accessible within the folded precursor8, and many peptide hormones have to be released from precursors to be perceived elsewhere by receptors29,30. The CSP, however, is very small, and much of the csp22 peptide is already exposed (Fig. 3d). In addition, since the csp22 peptide is at the N terminus, only C-terminal processing would be required for csp22 release. However, the timing of the oxidative burst upon CspD is not significantly different from that of csp22 (Figs. 1c,e and 2b), and we were unable to block CspD perception with subtilase inhibitors PMSF or Epi1 (Extended Data Fig. 7). And although csp22 binding to CORE has been demonstrated11, unprocessed CSPs are likely to bind to CORE, similar to how flagellin protein can bind FLS2 (ref. 31). The hypothesis that processing is not required for CSP perception is also consistent with the distribution of detected peptides over the CspD protein when incubated with AF as the detected peptide pattern suggests that there were only two initial processing sites (S1 and S2; Fig. 3b). Whereas processing at site S1 inactivates csp22, site S2 is far beyond the csp22 peptide. Furthermore, incubation of purified CspD with purified SBT5.2a indicates that site S1 is cleaved first and cleavage at S2 might be a secondary cleavage event (Fig. 6c). These observations indicate that processing might not be important for the release of the csp22 elicitor. By contrast, the inactivation of the csp22 epitope by SBT5.2 at site S1 seemed a more relevant process.

Indeed, subsequent experiments with csp22 confirmed that csp22 is unstable in the apoplast. We provided multiple lines of evidence that SBT5.2s are both required and sufficient for inactivating csp22. Inactivation of csp22 in AF can be blocked with SBT inhibitor Epi1, by silencing SBT5.2s or by inactivating SBT5.2s by genome editing. Conversely, purified SBT5.2a is sufficient to inactivate csp22 and can cleave the quenched octapeptide carrying the presumed S1 cleavage site. Also, the processing of CspD protein by purified SBT5.2a suggests that the S1 site is cleaved by SBT5.2. The S1 site in csp22 (VKWF ↓ NNAK) is consistent with the known promiscuity of this protease. SBT5.2 is also required for cleaving the prodomain of proRcr3 in the region carrying the sequence (EFKI-)NDLSDDYM(-PSN) irrespective of random mutagenesis in this region21, and purified SBT5.2a was found to cleave various recombinant proteins at sites TTLF ↓ GVPI, YNYY ↓ DFYD, YNYH ↓ YMDV, YHYM ↓ DVWG, TTVT ↓ VSSA, VTVS ↓ SAST, MFLE ↓ AIPM and AIPM ↓ SIPP20. Although there are often hydrophobic residues at positions P4, P2 and P1 (Val, Trp and Phe in csp22, respectively), there are several exceptions, indicating that SBT5.2 might be promiscuous and that it is rather the combination of residues in a peptide that may dictate SBT5.2 cleavability. The promiscuity of SBT5.2 is consistent with observations made for Arabidopsis SBT5.2, which was found to process the precursors of Inflorescence Deficient in Abscission (proIDA32) and Epidermal Patterning Factor (proEPF2; ref. 33), as well as flagellin34, at sites that do not share much homology.

The consequence of csp22 processing is that the csp22 levels are suppressed by SBT5.2 and that, consequently, the immune response is dampened by SBT5.2 s. Indeed, priming by low csp22 concentrations induces PTI only in sbt5.2 mutants (Fig. 7c), confirming that SBT5.2 dampens csp22-triggered PTI. A dampened immune response may benefit the plant by restricting costly immune responses to the site of pathogen infection by avoiding the diffusion of peptide elicitors. Likewise, csp22 degradation will also result in a more temporal immune response, so that PTI is no longer triggered in plants after an infection has cleared. Both temporal and spatial dampening of immune response would avoid unnecessary activation of costly immune responses and make more resources available to promote plant growth.

But the fact that SBT5.2s inactivate csp22 is also a clear benefit to the pathogen, and this implies that pathogens might take advantage of plant SBT5.2 to degrade their elicitors. The relevance of elicitor degradation is testified by the fact that P. syringae secretes AprA, a metalloprotease that inactivates flg22, the main elicitor from flagellin35. Selection for destabilization also occurred in Avr4, a protein secreted by the fungal tomato pathogen Cladosporium fulvum that is recognized by the Cf-4 immune receptor in tomato. Immune evasion resulted in virulent races that predominantly carry substitutions of Cys residues that destabilize Avr4 in the apoplast but maintain its ability to protect chitin against chitinases36,37. We were also able to demonstrate that SBT5.2 cleaves the flg22 epitope in flagellin, inactivating its immunogenicity38. Interestingly, the fact that both CSP and flagellin are cleaved first in the immunogenic epitopes instead of elsewhere is remarkable and might have resulted from evolutionary pressure to avoid recognition.

Interestingly, we detected some variation in the stability of the csp22 peptides, although all peptides are eventually degraded by SBT5.2. Since the tested peptides all carry the VKWF tetrapeptide before the cleavage site, the differential stability is probably caused by variation in residues after the cleavage site. This indicates that the NDEK tetrapeptide after the cleavage site reduces SBT5.2 sensitivity, whereas the NTSK sequence increases SBT5.2 sensitivity when compared with the NNAK tetrapeptide from CspD. These observations indicate that acidic residues (Asp or Glu) at the P2′ and P3′ positions may reduce the SBT5.2 sensitivity of csp22 peptides. The different stabilities of the csp22 peptides implies that the relative concentrations of these different csp22 peptides will change after infection and at a distance from the infection site.

A recent pangenomic study on the diversity of csp22 in bacteria revealed that csp22 exhibits significant copy and epitope variation39. Interestingly, 25% of the tested csp22 peptides were not immunogenic in tomato carrying the CORE receptor, and some of these peptides were found to suppress csp22-mediated PTI by blocking the perception of immunogenic csp22. Remarkably, these non-immunogenic csp22 peptides have even more variation surrounding the S1 site, indicating that antagonistic csp22 peptides might have an enhanced stability to increase their ability to interfere with PTI signalling. These predictions, however, are all based on assays with synthetic csp22 peptides. Although five CSP genes are expressed in PtoDC3000 during infection, and derived peptides have been detected in crude AFs from infected plants by proteomics39, it remains to be shown which CSPs are perceived during infection. Experiments to delete CSP genes or alter the csp22 sequence are challenging because CSPs are collectively essential for bacterial survival40,41, and the RNA-binding motif overlaps with the csp22 epitope.

SBT5.2 is the most active subtilase in the apoplast of N. benthamiana, and its role in degrading csp22 epitopes (this work) and flg22 epitopes38 has now been demonstrated. Without preincubation in AF, we did not detect an altered ROS burst upon csp22/flg22 signalling in the sbt5.2 mutant, indicating that SBT5.2 is not involved in signalling itself. We did also not detect any macroscopic developmental phenotype of sbt5.2 mutants23. Arabidopsis sbt5.2 mutants also grow normally32. Given its abundance and promiscuity, we hypothesize that SBT5.2 might mediate the removal of unstable proteins in the apoplast to maintain extracellular protein homeostasis. The identification of proteins accumulating in the apoplast of sbt5.2 mutant plants may therefore provide more insights into the endogenous role of SBT5.2.

The fact that a single bacterium produces multiple CSPs including csp22 peptides that have different stability and immunogenicity indicates that the outcome of interactions involving CSPs is complicated. These interactions will be further complicated by the variation in apoplastic proteases between plant species and upon the secretion of immune proteases. These interactions can be further fine-tuned by the secretion of protease inhibitors by pathogens. Although the csp22 sequences are evolutionarily constrained by the fact that they contain the RNA-binding motif required for the intrinsic function of CSP9, these observations indicate that csp22 variation might underlie a fascinating natural battlefield that is pivotal for the outcome of plant–bacterium interactions.

Nicotiana benthamiana plants (LAB genotype) were grown at 21 °C (night) and 22–23 °C (day) under a 16 h light (80–120 μmol m−2 s−1)/8 h dark routine in a greenhouse until use.

All the primers and plasmids used are summarized in Supplementary Tables 1 and 2, respectively. Expression vector pJK155 was generated by cloning inserts of pJP001, pJK120, pFGH029 and pJP002 into pJK082 using a BsaI Golden Gate reaction, resulting in pJK155 (pET28b–T7::OmpA–His–TEV–EPIC1). Open reading frames of CapB and CspD from PtoDC3000 were amplified from genomic DNA of PtoDC3000 using the primers listed in Supplementary Table 1 and cloned into pJK155, which was linearized by PCR with primers 5′-gcttggatccggctgctaac-3′ and 5′-accttggaagtataggttttcgtg-3′ using the Gibson ligation method, resulting in pCC03 (pET28b–T7::OmpA–His–TEV–CapB) and pCC04 (pET28b–T7::OmpA–His–TEV–CspD). The open reading frame of NbSBT5.2a was commercially synthesized with a C-terminal 6HIS tag (Twist Bioscience; Supplementary Table 1) and assembled with the Golden Gate–compatible vector pJK001c binary vector21, the 35S promoter module (pICH51288) and the 35S terminator module (pICH41414) in a BsaI reaction resulting in expression plasmid pPB097 for transient protein expression of SBT5.2a–His in N. benthamiana. This plasmid was transformed into E. coli DH10β for amplification, purified and then transformed into Agrobacterium tumefaciens GV3101(pMP90). Transformants were selected on plates of LB-agar medium containing 25 µg ml−1 rifampicin, 10 µg ml−1 gentamycin and 50 µg ml−1 kanamycin.

Plasmid pCC04 was transformed into the Rosetta strain of E. coli, protein expression was induced with 0.4 mM IPTG at 20 °C and proteins were purified on HisPur Ni-NTA Resin (Thermo Scientific) according to the manufacturer’s instructions. Protein purity was verified by protein gel electrophoresis followed by Coomassie staining and western blotting using anti-His (HRP) antibody (Miltenyi Biotec). Signals were generated by chemiluminescence using Clarity ECL Western Blotting Substrate (BioRad) and detected with the ImageQuant LAS 4000 (GE Healthcare). Purified proteins were further concentrated using an Amicon Ultra centrifugal filter device (3 kDa MW cut-off, Millipore). Protein quantity was measured using Bradford method (Sigma-Aldrich). Proteins were stored in aliquots at −80 °C until use.

To express proteins transiently in N. benthamiana by agroinfiltration, overnight cultures of A. tumefaciens GV3101(pMP90) carrying binary plasmids to express Epi1 (pFGH048; ref. 42) or SBT5.2a–HIS (pPB097) were harvested by centrifugation. Cells were resuspended in induction buffer (10 mM MgCl2, 10 mM MES pH 5.0 and 150 µM acetosyringone) and mixed (1:1) with agrobacteria carrying the silencing inhibitor P19 at OD600 = 0.5. After 1 h at 21 °C, the cells were infiltrated with a needleless syringe into the abaxial side of three leaves of four-week-old N. benthamiana. The leaves were harvested and processed at the indicated days after agroinfiltration.

AFs were collected as described previously43. Leaves from WT, agroinfiltrated or VIGS-silenced plants were submerged in ice-cold water and infiltrated by applying a vacuum for 5 min. The surface of water-infiltrated leaves was dried with absorbing paper, and the leaves were carefully mounted in an empty 20 ml syringe and placed in a 50 ml tube. AFs were collected by centrifugation at 2,000 g at 4 °C for 20 min and used immediately or flash-frozen and stored at −20 °C.

Four-week-old N. benthamiana leaves were infiltrated with a 1:1 mixture (final OD600 = 0.5 for each) of A. tumefaciens GV3101 containing the silencing suppressor P19 and pPB097 or pJP008. AF containing SBT5.2a–His or P69B–His was extracted six days after infiltration and purified as previously described44,45.

The ROS burst assay was performed as previously described43 except that L-012 (Wako Chemical) was used instead of luminol and the diameter of leaf discs used here was 4 mm rather than 6 mm. Briefly, after incubation in water overnight, one leaf disc (4 mm in diameter) was added to 100 µl of solution containing 25 ng µl−1 L-012, 25 ng µl−1 HRP and specified elicitor treatments. For assays with elicitors treated with AFs, 500 nM csp22 or purified CspD was incubated in AFs from the specified N. benthamiana leaves (WT, agroinfiltrated or VIGS-silenced) for one hour at room temperature with slight shaking. After incubation, 25 ng µl−1 L-012 and 25 ng µl−1 HRP were added to the AFs. Chemiluminescence was measured immediately with an Infinite M200 plate reader (Tecan) every minute for one hour.

All peptides used were custom-synthesized by GenScript and are summarized in Supplementary Table 3.

Qcsp8 (VKWFNNAK) from CspD was commercially synthesized with a DABCYL N-terminal modification and an EDANS C-terminal modification (GenScript) at a purity of 95.7%. It was resuspended in DMSO at a concentration of 1 mM. This stock solution was further diluted in water to a concentration of 200 µM. AFs or purified SBT5.2a or P69B were mixed with Qcsp8 at a final concentration of 10 µM in a volume of 100 µl, and fluorescence was measured immediately or after the indicated incubation time at 21 °C using an Infinite M200 plate reader (Tecan) with an excitation wavelength of 335 nm and an emission wavelength of 493 nm.

Purified CspD (stock concentration, 10 μM; final concentration, 2 μM) was incubated in AFs from the specified N. benthamiana leaves (WT, agroinfiltrated or VIGS-silenced) for the indicated times at room temperature with slight shaking. Proteins were analysed via SDS–PAGE and Coomassie staining.

First, 10 ng µl−1 of purified CspD protein produced in E. coli was incubated in AF from WT N. benthamiana for 15 min or 60 min at room temperature. CspD and AF alone were used as negative controls, and two technical replicates were included for each treatment or control. After incubation, samples for the analysis of endogenously digested peptides in the AF were generated by supplementing the AF with four volumes of MS-grade acetone, followed by incubation on ice for one hour and centrifugation at 18,000 g for 15 min. Four fifths of the supernatants were then transferred to fresh Eppendorf tubes, and the acetone was evaporated by vacuum centrifugation. The dried peptide samples were then dissolved in 0.1% formic acid and immediately analysed without further clean-up. LC–MS/MS analysis and peptide identification were performed as previously described43 except that MS/MS spectra data were searched against the sequence of CspD protein. Identified CspD peptides in samples of CspD incubated in AF were normalized by subtraction of the data generated from negative controls and aligned with the CspD protein sequence.

Each sample was analysed on an Orbitrap Elite instrument (Thermo)46 that was coupled to an EASY-nLC 1000 LC system (Thermo) and an Orbitrap Fusion Lumos (Thermo) coupled to an EASY-nLC 1200 LC system (Thermo). The LC systems were operated in the one-column mode. The analytical column was a fused silica capillary (75 µm × 46 cm) with an integrated fritted emitter (15 µm; CoAnn Technologies) packed in-house with Kinetex C18-XB core shell 1.7 µm resin (Phenomenex). The analytical column was encased by a column oven (Sonation) and attached to a nanospray flex ion source (Thermo). The column oven temperature was adjusted to 50 °C during data acquisition. The LC system was equipped with two mobile phases: solvent A (0.2% formic acid in water) and solvent B (0.2% formic acid, 19.8% water and 80% acetonitrile). All solvents were of UPLC grade (Honeywell). Peptides were directly loaded onto the analytical column with a maximum flow rate that would not exceed the set pressure limit of 980 bar (usually around 0.6–1.0 µl min−1). Peptides were subsequently separated on the analytical column by running a gradient of solvent A and solvent B.

RAW spectra were submitted to an Andromeda47 search in MaxQuant (v.2.0.2.0) using the default settings48. Label-free quantification and match-between-runs were activated49. The MS/MS spectra data were searched against a project-specific database containing two sequences of interest (ACE_0686_SOI_v01.fasta; two entries). All searches included a contaminants database search (as implemented in MaxQuant, 245 entries). The contaminants database contains known MS contaminants and was included to estimate the level of contamination. Andromeda searches allowed oxidation of methionine residues (16 Da) and acetylation of the protein N terminus (42 Da). No dynamic modifications were selected. Enzyme specificity was set to ‘unspecific’. The instrument type in Andromeda searches was set to Orbitrap, and the precursor mass tolerance was set to ±20 ppm (first search) and ±4.5 ppm (main search). The MS/MS match tolerance was set to ±0.5 Da. The peptide spectrum match false discovery rate and the protein false discovery rate were set to 0.01 (on the basis of the target-decoy approach). The minimum peptide length was 6 amino acids, and the maximum length was 36. For protein quantification, unique and razor peptides were allowed. Modified peptides were allowed for quantification. The minimum score for modified peptides was 40. Label-free protein quantification was switched on, and unique and razor peptides were considered for quantification with a minimum ratio count of 2. Retention times were recalibrated on the basis of the built-in nonlinear time-rescaling algorithm. MS/MS identifications were transferred between LC–MS/MS runs with the ‘match between runs’ option, in which the maximal match time window was set to 0.7 min and the alignment time window set to 20 min. The quantification is based on the ‘value at maximum’ of the extracted ion current. At least two quantitation events were required for a quantifiable protein. Further analysis and filtering of the results were done in Perseus v.1.6.10.0 (ref. 50). Comparison of protein group quantities (relative quantification) between different MS runs is based solely on the label-free quantifications as calculated by MaxQuant with the MaxLFQ algorithm49.

N. benthamiana plants silenced for NbCORE, SBT5.2, GUS (negative control) and phytoene desaturase (positive control) were generated using VIGS as previously described13,23. Briefly, overnight cultures of A. tumefaciens GV3101 were collected and resuspended in agroinfiltration buffer (10 mM MgCl2, 10 mM MES pH 5.0 and 100 mM acetosyringone). Suspensions of bacteria containing TRV2gg::NbCORE, TRV2::SBT5.2 and TRV2::GUS were mixed 1:1 separately with bacteria containing TRV1 at OD600 = 0.5 for each bacterium. After incubation for one hour at room temperature, the mixed cultures were infiltrated into true leaves of two-week-old N. benthamiana plants. The infiltrated seedlings were grown in a growth chamber until use.

FP-TAMRA (Thermo Scientific) was prepared as 10 µM stock solutions in dimethyl sulfoxide. Labelling was performed as described previously43. For fluorescence gel imaging, the AFs were incubated with 0.2 µM probes for 1 h at room temperature in the dark. The labelling reactions were stopped by adding 4× gel loading buffer (200 mM Tris-HCl (pH 6.8), 400 mM DTT, 8% SDS, 0.4% bromophenol blue and 40% glycerol) and heating at 90 °C for 5 min. The proteins were separated on SDS–PAGE, and fluorescence was detected from protein gels using the Typhoon FLA 9000 scanner (GE Healthcare Life Sciences) using Cy3 settings (532 nm excitation and 610PB filter).

For the infection assays, csp22 peptides were diluted in water. Three fully expanded leaves of three- to four-week-old N. benthamiana plants were infiltrated with different concentrations of csp22 peptide or with water as a mock control. 24 h later, infiltrated leaves were infiltrated with 105 CFU ml−1 PtoDC3000(ΔhQ) or Pta6605. The next day, three leaf discs were punched with a cork borer from each infected leaf and surface-sterilized with 15% hydrogen peroxide for 2 min. The leaf discs were then washed twice in MilliQ and dried under sterile conditions. The leaf discs were placed into a 1.5 ml safe-lock Eppendorf tube with three 3-mm-diameter metal beads and 1 ml of MilliQ. The tubes were placed in tissue lyser for 5 min at 30 Hertz per second. 20 µl of undiluted tissue and serial dilutions were plated on LB-agar plates containing Pseudomonas CFC Agar Supplement (Oxoid SR0103). The plates were allowed to dry and incubated at 28 °C for two days, and then colonies were counted. P values were calculated using two-tailed Student’s t-tests to compare bacterial growth between leaves from WT and sbt5.2 mutant plants.

All values shown are mean values, and the error intervals shown represent the standard error of the mean, unless otherwise indicated. All experiments have been reproduced, and representative datasets are shown.

Proteomes of P. syringae strains were obtained from the pseudomonas.com database51, and proteomes of Ralstonia solanacearum and Xanthomonas campestris were obtained from the RefSeq database52. Cold-shock-domain-containing proteins were identified using the hmmsearch function from HMMER v.3.3.2 (ref. 53) with the PF00313 (cold-shock DNA-binding domain) Pfam profile54. Only sequences with a recognizable csp22 epitope were retained. Protein sequences were aligned using MAFFT v.7 with the L-INS-i algorithm55. A maximum likelihood phylogenetic tree was constructed using IQ-TREE v.2 (ref. 56) with the best-fit substitution model from ModelFinder57. Branch support values were calculated using ultrafast bootstrap with 1,000 replications. Phylogenetic trees were visualized using iTOL58 with midpoint rooting.

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

The MS proteomics data for the on-bead digestions have been deposited to the ProteomeXchange Consortium via the PRIDE59 partner repository (https://www.ebi.ac.uk/pride/archive/) with the dataset identifier PXD048912. Source data are provided with this paper.

Boller, T. & Felix, G. A renaissance of elicitors: perception of microbe-associated molecular patterns and danger signals by pattern-recognition receptors. Annu. Rev. Plant Biol. 60, 379–406 (2009).

Article CAS PubMed Google Scholar

Couto, D. & Zipfel, C. Regulation of pattern recognition receptor signalling in plants. Nat. Rev. Immunol. 16, 537–552 (2016).

Article CAS PubMed Google Scholar

Jones, J. D. G. & Dangl, J. L. The plant immune system. Nature 444, 323–329 (2006).

Article CAS PubMed Google Scholar

Van der Hoorn, R. A. L. Plant proteases: from phenotypes to molecular mechanisms. Annu. Rev. Plant Biol. 59, 191–223 (2008).

Article PubMed Google Scholar

Godson, A. & Van der Hoorn, R. A. L. The front line of defence: a meta-analysis of apoplastic proteases in plant immunity. J. Exp. Bot. 72, 3381–3394 (2021).

Article CAS PubMed PubMed Central Google Scholar

Mott, G. A., Middleton, M. A., Desveaux, D. & Guttman, D. S. Peptides and small molecules of the plant-pathogen apoplastic arena. Front Plant Sci. 5, 677 (2014).

Article PubMed PubMed Central Google Scholar

Boller, T. & He, S. Y. Innate immunity in plants: an arms race between pattern recognition receptors in plants and effectors in microbial pathogens. Science 324, 742–744 (2009).

Article CAS PubMed PubMed Central Google Scholar

Chen, C., van der Hoorn, R. A. L. & Buscaill, P. Releasing hidden MAMPs from precursor proteins in plants. Trends Plant Sci. 29, 428–436 (2023).

Article PubMed Google Scholar

Felix, G. & Boller, T. Molecular sensing of bacteria in plants—the highly conserved RNA-binding motif RNP-1 of bacterial cold shock proteins is recognized as an elicitor signal in tobacco. J. Biol. Chem. 278, 6201–6208 (2003).

Article CAS PubMed Google Scholar

Saur, I. M. et al. NbCSPR underlies age-dependent immune responses to bacterial cold shock protein in Nicotiana benthamiana. Proc. Natl Acad. Sci. USA 113, 3389–3394 (2016).

Article CAS PubMed PubMed Central Google Scholar

Wang, L. et al. The pattern-recognition receptor CORE of Solanaceae detects bacterial cold-shock protein. Nat. Plants 2, 16185 (2016).

Article CAS PubMed Google Scholar

Bae, W., Xia, B., Inouye, M. & Severinov, K. Escherichia coli CspA-family RNA chaperones are transcription antiterminators. Proc. Natl Acad. Sci. USA 97, 7784–7789 (2000).

Article CAS PubMed PubMed Central Google Scholar

Dodds, I., Chen, C., Buscaill, P. & Van der Hoorn, R. A. L. Depletion of the NbCORE receptor drastically improves agroinfiltration productivity in older Nicotiana benthamiana plants. Plant Biotechnol. J. 21, 1103–1105 (2023).

Article CAS PubMed PubMed Central Google Scholar

Nie, J. et al. A receptor-like protein from Nicotiana benthamiana mediates VmE02 PAMP-triggered immunity. New Phytol. 229, 2260–2272 (2021).

Article CAS PubMed Google Scholar

Yang, Y. et al. Convergent evolution of plant pattern recognition receptors sensing cysteine-rich patterns from three microbial kingdoms. Nat. Commun. 14, 3621 (2023).

Article CAS PubMed PubMed Central Google Scholar

Nobori, T. et al. Transcriptome landscape of a bacterial pathogen under plant immunity. Proc. Natl Acad. Sci. USA 115, E3055–E3064 (2018).

Article CAS PubMed PubMed Central Google Scholar

Grosse-Holz, F. et al. The transcriptome, extracellular proteome and active secretome of agroinfiltrated Nicotiana benthamiana uncover a large, diverse protease repertoire. Plant Biotechnol. J. 16, 1068–1084 (2018).

Article CAS PubMed Google Scholar

Tian, M., Huitema, E., Da Cunha, L., Torto-Alalibo, T. & Kamoun, S. A Kazal-like extracellular serine protease inhibitor from Phytophthora infestans targets the tomato pathogenesis-related protease P69B. J. Biol. Chem. 279, 26370–26377 (2004).

Article CAS PubMed Google Scholar

Jutras, P. V. et al. Activity-based proteomics reveals nine target proteases for the recombinant protein-stabilizing inhibitor SlCYS8 in Nicotiana benthamiana. Plant Biotechnol. J. 17, 1670–1678 (2019).

Article CAS PubMed PubMed Central Google Scholar

Puchol Tarazona, A. A. et al. Identification of two subtilisin-like serine proteases engaged in the degradation of recombinant proteins in Nicotiana benthamiana. FEBS Lett. 595, 379–388 (2021).

Article CAS PubMed Google Scholar

Paulus, J. K. et al. Extracellular proteolytic cascade in tomato activates immune protease Rcr3. Proc. Natl Acad. Sci. USA 117, 17409–17417 (2020).

Article CAS PubMed PubMed Central Google Scholar

Sueldo, D. J. et al. Activity-based proteomics uncovers suppressed hydrolases and a neo-functionalised antibacterial enzyme at the plant–pathogen interface. New Phytol. 241, 394–408 (2024).

Article CAS PubMed Google Scholar

Beritza, K. et al. SBT5.2s are the major active extracellular subtilases processing IgG antibody 2F5 in the N. benthamiana apoplast. Plant Biotechnol. J. (in the press).

Kourelis, J. et al. A homology-guided, genome-based proteome for improved proteomics in the alloploid Nicotiana benthamiana. BMC Genomics 20, 722 (2019).

Article PubMed PubMed Central Google Scholar

Ranawaka, B. et al. A multi-omic Nicotiana benthamiana resource for fundamental research and biotechnology. Nat. Plants 9, 1558–1571 (2023).

Article CAS PubMed PubMed Central Google Scholar

Kvitko, B. H. et al. Deletions in the repertoire of Pseudomonas syringae pv. tomato DC3000 type III secretion effector genes reveal functional overlap among effectors. PLoS Pathog. 5, e1000388 (2009).

Article PubMed PubMed Central Google Scholar

Zipfel, C. et al. Bacterial disease resistance in Arabidopsis through flagellin perception. Nature 428, 764–767 (2004).

Article CAS PubMed Google Scholar

Shimizu, R. et al. The DeltafliD mutant of Pseudomonas syringae pv. tabaci, which secretes flagellin monomers, induces a strong hypersensitive reaction (HR) in non-host tomato cells. Mol. Genet. Genomics 269, 21–30 (2003).

Article CAS PubMed Google Scholar

Stintzi, A. & Schaller, A. Biogenesis of post-translationally modified peptide signals for plant reproductive development. Curr. Opin. Plant Biol. 69, 102274 (2022).

Article CAS PubMed Google Scholar

Yang, H. et al. Subtilase-mediated biogenesis of the expanded family of SERINE RICH ENDOGENOUS PEPTIDES. Nat. Plants 9, 2085–2094 (2023).

Article CAS PubMed Google Scholar

Meindl, T., Boller, T. & Felix, G. The bacterial elicitor flagellin activates its receptor in tomato cells according to the address-message concept. Plant Cell 12, 1783–1794 (2000).

CAS PubMed PubMed Central Google Scholar

Schardon, K. et al. Precursor processing for plant peptide hormone maturation by subtilisin-like serine proteinases. Science 354, 1594–1597 (2016).

Article CAS PubMed Google Scholar

Engineer, C. B. et al. Carbonic anhydrases, EPF2 and a novel protease mediate CO2 control of stomatal development. Nature 513, 246–250 (2014).

Article CAS PubMed PubMed Central Google Scholar

Matsui, S. et al. Arabidopsis SBT5.2 and SBT1.7 subtilases mediate C-terminal cleavage of flg22 epitope from bacterial flagellin. Nat. Commun. 15, 3762 (2024).

Article CAS PubMed PubMed Central Google Scholar

Pel, M. J. et al. Pseudomonas syringae evades host immunity by degrading flagellin monomers with alkaline protease AprA. Mol. Plant Microbe Interact. 27, 603–610 (2014).

Article CAS PubMed Google Scholar

Joosten, M. H. A. J., Vogelsang, R., Cozijnsen, T. J., Verberne, M. C. & De Wit, P. J. G. M. The biotrophic fungus Cladosporium fulvum circumvents Cf-4-mediated resistance by producing unstable AVR4 elicitors. Plant Cell 9, 367–379 (1997).

CAS PubMed PubMed Central Google Scholar

van den Burg, H. A. et al. Natural disulfide bond-disrupted mutants of AVR4 of the tomato pathogen Cladosporium fulvum are sensitive to proteolysis, circumvent Cf-4-mediated resistance, but retain their chitin binding ability. J. Biol. Chem. 278, 27340–27346 (2003).

Article PubMed Google Scholar

Buscaill, P. et al. Subtilase SBT5.2 counterbalances flagellin immunogenicity in the plant apoplast. Preprint at bioRxiv https://doi.org/10.1101/2024.05.23.595557 (2024).

Stevens, D. M. et al. Natural variation of immune epitopes reveals intrabacterial antagonism. Proc. Natl Acad. Sci. USA 121, e2319499121 (2024).

Article CAS PubMed PubMed Central Google Scholar

Xia, B., Ke, H. & Inouye, M. Acquirement of cold sensitivity by quadruple deletion of the cspA family and its suppression by PNPase S1 domain in Escherichia coli. Mol. Microbiol. 40, 179–188 (2001).

Article CAS PubMed Google Scholar

Graumann, P., Wendrich, T. M., Weber, M. H., Schröder, K. & Marahiel, M. A. A family of cold shock proteins in Bacillus subtilis is essential for cellular growth and for efficient protein synthesis at optimal and low temperatures. Mol. Microbiol. 25, 741–756 (1997).

Article CAS PubMed Google Scholar

Grosse-Holz, F. et al. Three unrelated protease inhibitors enhance accumulation of pharmaceutical recombinant proteins in Nicotiana benthamiana. Plant Biotechnol. J. 16, 1797–1810 (2018).

Article CAS PubMed PubMed Central Google Scholar

Buscaill, P. et al. Glycosidase and glycan polymorphism control hydrolytic release of immunogenic flagellin peptides. Science 364, eaav0748 (2019).

Article CAS PubMed Google Scholar

Schuster, M., Paulus, J. K., Kourelis, J. & Van der Hoorn, R. A. L. Purification of His-tagged proteases from the apoplast of agroinfiltrated N. benthamiana. Methods Mol. Biol. 2447, 53–66 (2022).

Article CAS PubMed Google Scholar

Homma, F., Huang, J. & van der Hoorn, R. A. L. AlphaFold-Multimer predicts cross-kingdom interactions at the plant–pathogen interface. Nat. Commun. 14, 6040 (2023).

Article CAS PubMed PubMed Central Google Scholar

Michalski, A. et al. Ultra high resolution linear ion trap Orbitrap mass spectrometer (Orbitrap Elite) facilitates top down LC MS/MS and versatile peptide fragmentation modes. Mol. Cell. Proteomics 11, O111.013698 (2012).

Article PubMed Google Scholar

Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

Article CAS PubMed Google Scholar

Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

Article CAS PubMed Google Scholar

Cox, J. et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell. Proteomics 13, 2513–2526 (2014).

Article CAS PubMed PubMed Central Google Scholar

Tyanova, S. et al. The PERSEUS computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

Article CAS PubMed Google Scholar

Winsor, G. L. et al. Enhanced annotations and features for comparing thousands of Pseudomonas genomes in the Pseudomonas genome database. Nucleic Acids Res. 44, D646–D653 (2016).

Article CAS PubMed Google Scholar

O'Leary, N. A. et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 44, D733–D745 (2016).

Article CAS PubMed Google Scholar

Finn, R. D., Clements, J. & Eddy, S. R. HMMER web server: interactive sequence similarity searching. Nucleic Acids Res. 39, W29–W37 (2011).

Article CAS PubMed PubMed Central Google Scholar

Mistry, J. et al. Pfam: the protein families database in 2021. Nucleic Acids Res. 49, D412–D419 (2021).

Article CAS PubMed Google Scholar

Katoh, K. & Standley, D. M. MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol. Biol. Evol. 30, 772–780 (2013).

Article CAS PubMed PubMed Central Google Scholar

Minh, B. Q. et al. IQ-TREE 2: new models and efficient methods for phylogenetic inference in the genomic era. Mol. Biol. Evol. 37, 1530–1534 (2020).

Article CAS PubMed PubMed Central Google Scholar

Kalyaanamoorthy, S., Minh, B. Q., Wong, T. K. F., von Haeseler, A. & Jermiin, L. S. ModelFinder: fast model selection for accurate phylogenetic estimates. Nat. Methods 14, 587–589 (2017).

Article CAS PubMed PubMed Central Google Scholar

Letunic, I. & Bork, P. Interactive Tree Of Life (iTOL) v5: an online tool for phylogenetic tree display and annotation. Nucleic Acids Res. 49, W293–W296 (2021).

Article CAS PubMed PubMed Central Google Scholar

Vizcaíno, J. A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, 11033 (2016).

Article PubMed PubMed Central Google Scholar

Download references

We thank J. Kourelis for cloning pJK155; U. Pyzio for excellent plant care; and S. Rodgers, P. Bowman and C. O’Brien for excellent technical support. This project was financially supported by the National Key Research and Development Projects no. 2022YFE0199500 and the China Scholarship Council (C.C.), BBSRC 17RM3 project no. BB/R017913/3 ‘GH35’ (P.B.) and 19RM3 project no. BB/015128/1 ‘Galactosyrin’ (P.B. and N.S.), and ERC-2020-AdG project no. 101019324 ‘ExtraImmune’ (J.H. and R.A.L.v.d.H.).

Institute of Biotechnology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China

Changlong Chen

Plant Chemetics Laboratory, Department of Biology, University of Oxford, Oxford, UK

Changlong Chen, Pierre Buscaill, Nattapong Sanguankiattichai, Jie Huang & Renier A. L. van der Hoorn

ZMB Chemical Biology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany

Farnusch Kaschani & Markus Kaiser

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

C.C. performed most of the experiments. P.B. supervised the project and performed the infection assays and Qcsp8 experiments. F.K. and M.K. generated the proteomics data. J.H. purified SBT5.2 and performed the CspD cleavage assay. N.S. performed the ROS assays presented in Figs. 7 and 8. R.A.L.v.d.H. conceived and supervised the project. R.A.L.v.d.H. and C.C. wrote the article with input from all co-authors.

Correspondence to Renier A. L. van der Hoorn.

The authors declare no competing interests.

Nature Plants thanks Jack Rhodes and Andreas Schaller for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

CapB and CspD with expressed with N-terminal His-TEV peptides in E. coli and purified over Ni-NTA. I, input sample; W1, wash with 100 mM imidazole; W2, wash with 150 mM imidazole; E, eluate with 250 mM imidazole. Samples were separated by SDS-PAGE and stained with Coomassie. This experiment was repeated two times with similar results.

Coverages of detected CspD peptides after incubation of CspD in water (bottom) or AF (top) for 15 (left) and 60 (right) minutes.

SBT5.2a-His was transiently expressed by agroinfiltration of N. benthamiana. AF was isolated at 6 dpi and purified over Ni-NTA. The column was washed with 50 mM imidazole and eluted with 250 mM imidazole. This experiment was repeated four times with similar results.

a, Leaf discs of 7-week old plants were treated with 0, 1 or 10 μM csp22 and luminescence with luminol was measured for one hour in relative light units (RLU). Error shades represent the STDEV of n = 4 replicates. b, Leaf discs of 7-week old plants were treated with 0, 1 or 10 μM csp22 and luminescence with LO-12 was measured for one hour in relative light units (RLU). Error shades represent the SE of n = 6 replicates.

Leaves of WT and sbt5.2 mutant N. benthamiana were infiltrated with water or 0.5 or 5 μM csp22 and incubated for 24 hours and then injected with 1 × 105 bacteria/mL of the flagellin mutant strain of Pseudomonas syringae pv. tabaci 6605 (Pta6605ΔfliC). Colony forming units (CFUs) were determined one day post infection (dpi). Error bars represent SE of n = 6 replicates.

Leaf discs of 6-week old plants were treated with 500 nM csp22 of PSPTO3984 or PSPTO4145, and ROS was measured in relative light units (RLU). Error shades represent the SE of n = 6 replicates.

a, Leaf discs of N. benthamiana were treated with 0.5 mM PMSF or 1% DMSO when floating on luminol-HRP solution and were triggered with 500 nM CspD protein and the release of reactive oxygen species (ROS) was detected by luminescence. Error shades represent SE of n = 6 replicates. b, Leaf discs taken from agroinfiltrated leaves expressing Epi1 or empty vector (EV) control, taken at 3 dpi, floating on luminol-HRP solution, were triggered with 500 nM CspD protein and the release of reactive oxygen species (ROS) was detected by luminescence. Error shades represent SE of n = 6 replicates.

Supplementary Tables 1–3.

Uncropped gel for Fig. 3a.

Uncropped gel for Fig. 5b,e.

Uncropped gel for Fig. 6a,c.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

Chen, C., Buscaill, P., Sanguankiattichai, N. et al. Extracellular plant subtilases dampen cold-shock peptide elicitor levels. Nat. Plants (2024). https://doi.org/10.1038/s41477-024-01815-8

Download citation

Received: 01 February 2024

Accepted: 05 September 2024

Published: 11 October 2024

DOI: https://doi.org/10.1038/s41477-024-01815-8

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative