AI-2 induces c-di-GMP synthesis by acting as an activator of YeaJ in S. Typhimurium
Although AI-2 is the well-known QS molecule produced by enteric bacteria including E. coli and S. Typhimurium7,26, its physiological role in these bacteria remains poorly understood. Consistent with several previous studies27,28, we found that deletion of luxS led to significantly reduced biofilm formation in S. Typhimurium (Supplementary Fig. 1a). However, deletion of the lsrB gene that encodes the only known AI-2 receptor in S. Typhimurium26 did not affect its ability to form biofilm, whereas the double mutant ΔlsrBΔluxS showed significantly decreased biofilm formation compared with the ΔlsrB mutant (Fig. 1a). Moreover, deletion of luxS led to enhanced swimming motility in S. Typhimurium with or without the native lsrB gene (Supplementary Fig. 1b and Fig. 1b). These results suggest that AI-2 could play a role in the motile-sessile transition independent of its receptor LsrB.
Given that a recent study has found that dCache_1 domain-containing AI-2 receptors are widely distributed in bacteria7, we investigated whether this type of AI-2 receptor is present in S. Typhimurium. Domain annotations of all protein sequences of S. Typhimurium by hmmscan program in HMMER (https://www.ebi.ac.uk/Tools/hmmer/search/hmmscan) showed that none of the protein sequences has the dCache_1 domain model (PF02743) as the best hit. Nevertheless, periplasmic ligand-binding domains (LBDs) of 5 transmembrane proteins DpiB (STM0625), YeaJ (STM1283), YedQ (STM1987), DcuS (STM4304) and CreC (STM4589) were found to hit the dCache_1 domain model with an E-value <1E-3. We thus examined whether these LBDs have the capacity to bind AI-2. In the Vibrio harveyi MM32 reporter assay, AI-2 binding activity was observed only for the LBD of YeaJ, but not for the LBDs of the other 4 proteins (Fig. 1c). Furthermore, the binding analysis by isothermal titration calorimetry (ITC) showed that the YeaJ-LBD binds AI-2 with a disassociation constant (Kd) value of 0.15 ± 0.02 μM (Fig. 1d). These results indicate that AI-2 is a high-affinity ligand for YeaJ.
YeaJ has been shown to be an active DGC that is involved in the regulation of motile-sessile transition in E. coli and S. Typhimurium29,30. By the in vitro DGC activity assay, we found that DPD/AI-2 stimulates the activity of YeaJ in synthesizing c-di-GMP (Fig. 1e and Supplementary Fig. 2). Consistent with this finding, when S. Typhimurium strains were cultured to the mid-exponential phase where the extracellular AI-2 activity reached the maximal level in the wild-type strain31 (Supplementary Fig. 3a), intracellular c-di-GMP level in the ΔluxS mutant was significantly lower than that in the wild type (Fig. 1f). Such reduction was partially restored by complementation with a plasmid encoding luxS (Fig. 1f), whereas the exogenous addition of DPD/AI-2 in cultures of ΔluxS resulted in a significant increase in intracellular c-di-GMP concentration (Fig. 1g). In contrast, deletion of luxS in the ΔyeaJ mutant did not lead to significant changes in the intracellular level of c-di-GMP (Supplementary Fig. 4), while the addition of DPD/AI-2 did not increase intracellular c-di-GMP concentration in ΔyeaJΔluxS (Fig. 1g). Consistent with previous studies29,30, ΔyeaJ showed reduced biofilm formation but enhanced motility compared with the wild type, whereas no significant difference in these phenotypes was observed between ΔyeaJΔluxS and ΔyeaJ (Supplementary Fig. 5a, b), suggesting that AI-2 regulates biofilm formation and motility via YeaJ. Collectively, these results indicate that AI-2 positively regulates intracellular c-di-GMP levels in S. Typhimurium by acting as an activator of YeaJ.
YeaJ homologs that sense AI-2 are also present in E. coli and other members of Enterobacterales
While matching the dCache_1 domain model at a less stringent E-value (1.8E-4), the LBD of YeaJ had the GAPES1 model (PF17155) as the best hit (E-value = 4.5E-153) in hmmscan searches (Fig. 2a). BLASTP searching of the National Center for Biotechnology Information (NCBI) non-redundant protein database followed by domain predictions using InterProScan 5 against the Pfam database showed that YeaJ homologs that possess a putative N-terminal GAPES1 domain and a putative C-terminal GGDEF domain are mainly distributed in the order Enterobacterales, including members of the families Enterobacteriaceae, Pectobacteriaceae and Hafniaceae (Supplementary Data 1 and Supplementary Fig. 6). To examine the ability of the GAPES1 domains of these YeaJ homologs to bind AI-2, we randomly selected one YeaJ homolog from each genus and prepared recombinant His6-GAPES1 proteins from the luxS+ E. coli strain. In the V. harveyi MM32 reporter assay, AI-2 binding activity was observed for the GAPES1 domains of YeaJ homologs from EHEC O157:H7 and 15 species from other genera (Fig. 2b). In contrast, no such activity was detected in the GAPES1 domains of two YeaJ homologs (KMK13526 and WP_034494714) from 2 species belonging to the genera Pluralibacter and Buttiauxella (Fig. 2b). Binding analysis by ITC showed that the GAPES1 domain of the YeaJ homolog from EHEC O157:H7 binds AI-2 with high affinity (Supplementary Fig. 7a). These data suggest that GAPES1 is a new type of extracytoplasmic sensor recognizing the AI-2 signal and YeaJ homologs whose DGC activities can be regulated by AI-2 are widespread among members of Enterobacterales.
By amino acid sequence alignment of the GAPES1 domains that have been tested for AI-2 binding activity, we found that two residues corresponding to Y210 and D239 of YeaJ are conserved in AI-2-binding GAPES1 domains but not in the two GAPES1 domains with no AI-2 binding activity (Fig. 2c). Mutations in each of these two residues resulted in marked reduction in AI-2 binding affinity for YeaJ-LBD (Fig. 1d and Fig. 2d). Furthermore, mutations of both non-conserved residues to conserved residues within KMK13526-LBD (H216Y/Q245D) and WP_034494714-LBD (Q208Y/E237D) increased their AI-2 binding affinity to levels (0.16-0.21 μM) (Supplementary Fig. 7b-e) that were comparable to that of YeaJ-LBD (Fig. 1d). These results suggest that the two highly conserved positions of the GAPES1 domains corresponding to Y210 and D239 of YeaJ may be key residues for AI-2 binding.
AI-2 negatively controls the T3SS-1 and attenuates the virulence of S. Typhimurium in infection via YeaJ
High levels of c-di-GMP in S. Typhimurium have been shown to reduce secretion of T3SS-1 effectors as well as invasion of epithelial cells23-25. As expected, when grown under Salmonella pathogenicity island 1 (SPI-1) inducing conditions32 to the mid-exponential phase (Supplementary Fig. 3b), deletion of luxS or yeaJ significantly promoted intracellular accumulation and secretion of the T3SS-1 effectors SipB and SopB (Fig. 3a). Such induction was abolished by the expression of luxS and yeaJ in the corresponding mutants (Fig. 3a). However, the double mutant ΔyeaJΔluxS produced and secreted SipB and SopB at levels similar to those by ΔyeaJ (Fig. 3a). These results suggest that AI-2 negatively regulates the production and secretion of T3SS-1 effectors through YeaJ.
We then investigated whether the rise in protein levels of SipB and SopB in the mutants ΔluxS and ΔyeaJ is due to increased expression of sipB and sopB at the transcriptional levels. Quantitative real-time PCR (qRT-PCR) analysis showed that the mRNA levels of sipB and sopB were significantly higher in ΔluxS and ΔyeaJ compared to the wild-type and complemented strains (Fig. 3b). Similar observations were made for the expression of sopE2 (Fig. 3b), which encodes a T3SS-1 effector whose secretion is also regulated by c-di-GMP signaling23. Moreover, promoter reporter assays showed that deletion of luxS or yeaJ led to significantly increased promoter activities of sopB, sopE2 and the sicAsipBCDA operon (Fig. 3c). These results indicate that AI-2-induced elevated c-di-GMP inhibits transcription of sopB, sopE2 and the sicAsipBCDA operon.
We further investigated the ability of S. Typhimurium strains to adhere to and invade human colonic epithelial Caco-2 cells. In contrast to the wild-type parent strain, mutants lacking luxS or yeaJ showed slightly enhanced adherence to (Fig. 3d), and significantly increased invasion of Caco-2 cells (Fig. 3e). Complementation returned their adherence and invasion ability to wild-type levels (Fig. 3d, e). However, in contrast to our results, secretion of T3SS-1 effectors and the ability to invade epithelial cells were not altered in ΔluxS compared to the wild type in a previous study by Perrett et al.33. We note that different culture conditions were used for both assays in the two studies. In our study, both assays were performed using cultures grown in modified LB medium containing 0.3 M NaCl without agitation (a condition for induction of the T3SS-1 encoded on SPI-132,34) to mid-exponential phase, when the AI-2 activity in the culture supernatant of the wild-type strain was maximal (Supplementary Fig. 3b), while the study by Perrett et al.33 used shaking cultures in normal LB medium with an OD600 of 1.0 for T3SS-1 secretion assays and in late log phase for invasion assays. We also found no differences between the wild type and ΔluxS with respect to their ability to invade epithelial cells in the conditions that Perrett et al.33 used (Supplementary Fig. 8). Thus, the discrepancy observed in LuxS regulation of the T3SS-1 and invasion of epithelial cells can be explained by the use of different culture conditions.
To further investigate whether deletion of luxS or yeaJ affects intestinal colonization after infection by a natural route, we performed competitive oral infections of streptomycin-treated BALB/c mice with an equal mixture of wild-type and mutant strains of S. Typhimurium. Competition assays showed that ΔluxS, ΔyeaJ and ΔyeaJΔluxS outcompeted the wild type ~2 to 3-fold, ~8 to 27-fold and ~8 to 28-fold, respectively (Fig. 3f). Competitive indexes between ΔluxS and the wild type, although drastically lower than those between ΔyeaJ and the wild type, are statistically different from a control competition assay between two derivatives of the wild-type SL1344 carrying the kanamycin-resistant pKT100 and the chloramphenicol-resistant pBBR1MCS1, respectively (Fig. 3f). In contrast, when ΔyeaJΔluxS competed against ΔyeaJ, these two mutants were recovered at similar levels in the small intestine, cecum, and feces (Fig. 3f). Similar results were also observed when competitions were conducted using the same strains with swapped antibiotic markers (Supplementary Fig. 9). These data suggest that AI-2 negatively regulates S. Typhimurium intestinal colonization via YeaJ.
We also evaluated the lethality of S. Typhimurium strains in BALB/c mice. In an oral infection model, ΔluxS and ΔyeaJ led to significantly increased mouse mortality compared to the wild-type strain, whereas infections with the mutants ΔyeaJΔluxS and ΔyeaJ produced similar mortality (Fig. 3g). Consistent with the role of SPI-1 in intestinal infection35,36, deletion of the gene that encodes the T3SS-1 ATPase InvC resulted in decreased mortality of mice after oral challenge (Fig. 3g). Moreover, mice infected with ΔinvCΔluxS and ΔinvCΔyeaJ showed similar mortality compared to those infected with ΔinvC (Fig. 3g), indicating that AI-2-mediated c-di-GMP signaling regulates the virulence of S. Typhimurium via T3SS-1. However, deletion of luxS or yeaJ did not affect virulence of S. Typhimurium after intraperitoneal inoculation (Supplementary Fig. 10), suggesting that AI-2-induced repression of the T3SS-1 via YeaJ has no major impact on systemic infection. Together, these results indicate that AI-2 exerts a negative regulatory effect on S. Typhimurium virulence during intestinal infection through modulating the function of the T3SS-1 via YeaJ.
Bile salts stimulate the DGC activity of YedQ to repress the T3SS-1 in S. Typhimurium
Bile, a major host-produced heterogeneous mixture of compounds encountered by bacteria in the small intestine, was previously shown to repress the expression of invasion-related genes within SPI-1 in S. Typhimurium37,38, but the mechanism of such regulation is poorly understood. While bile has been reported to increase intracellular c-di-GMP levels in V. cholerae10,14, our results showed that AI-2 induces transcriptional repression of T3SS-1 genes via c-di-GMP signaling (Fig. 3b, c), leading us to speculate that bile salts may modulate intracellular c-di-GMP levels to repress the T3SS-1 in S. Typhimurium. Indeed, when strain SL1344 was stimulated by porcine bile salts at a concentration (0.05%, w/v) comparable to that physiologically occurring in intestinal contents39, the cellular concentration of c-di-GMP increased approximately 5-fold (Fig. 4a). As expected, the promoter activities of sopB, sopE2, and sicAsipBCDA in the wild-type strain were significantly repressed following exposure to 0.05% bile salts (Fig. 4b). These results suggest that bile salts repress T3SS-1 gene expression via increasing intracellular c-di-GMP levels in S. Typhimurium.
Consistent with the known role of c-di-GMP in promoting biofilm formation23,29, the addition of 0.05% bile salts in cultures of strain SL1344 resulted in a significant increase in biofilm production (Fig. 4c). We further tested 9 individual components of bile salts to determine their contributions to biofilm formation. Intriguingly, the addition of taurocholate and taurodeoxycholate significantly stimulated biofilm formation, while the remaining 7 components of bile have no such effect (Fig. 4c). To identify CMEs that robustly respond to bile salts, we deleted each of the 17 genes encoding predicted CMEs6 and examined the ability of the mutants to form biofilms in response to bile salts and the individual bile components taurocholate and taurodeoxycholate. Whereas most of these mutations did not affect the response of S. Typhimurium to bile salts, the deletion of yedQ completely abrogated the bile-induced enhancement of biofilm formation (Fig. 4d). Moreover, the inclusion of 1 μM taurocholate or taurodeoxycholate in cultures of the wild-type strain resulted in a significant increase in intracellular c-di-GMP levels, while such induction was completely abolished in ΔyedQ (Fig. 4e). Complementation of the mutant with a plasmid derived copy of yedQ restored c-di-GMP modulation in response to taurocholate and taurodeoxycholate (Fig. 4e). These observations indicate that the bile components taurocholate and taurodeoxycholate stimulate an increase in intracellular c-di-GMP concentrations via the DGC YedQ.
We further examined whether the LBD of YedQ directly interact with taurocholate and taurodeoxycholate. Binding analysis by ITC showed that taurocholate and taurodeoxycholate bind to YedQ-LBD with Kd values of 0.17 ± 0.03 μM and 0.14 ± 0.02 μM, respectively (Fig. 4f). We then predicted the 3D structure of YedQ-LBD by Alphafold240 and performed a docking simulation to analyze the interaction between YedQ-LBD and taurocholate. The best docking conformation obtained by AutoDock Vina 1.1.241 suggests that taurocholate is inserted into a big cavity of YedQ-LBD (Fig. 4g). This conformation suggests that taurocholate makes close contact with D233, K236, Q264, L288, L289, D291, E295 and Q297 in the cavity (Fig. 4h). In support of this binding model, mutations in D233, K236, Q264, L288 and L289 drastically reduced the binding affinity of YedQ-LBD for taurocholate (Fig. 4i and Supplementary Fig. 11). Furthermore, taurocholate and taurodeoxycholate were able to induce the DGC activity of YedQ in c-di-GMP synthesis (Fig. 4j and Supplementary Fig. 12). These results indicate that taurocholate and taurodeoxycholate can induce intracellular accumulation of c-di-GMP in S. Typhimurium by directly engaging YedQ.
Next, we tested whether the expression of T3SS-1 genes is affected by the presence of taurocholate and taurodeoxycholate. Addition of 1 μM taurocholate or taurodeoxycholate strongly repressed the promoter activities of sopB, sopE2, and sicAsipBCDA in the wild-type strain, whereas this effect was completely abolished in the ΔyedQ mutant (Fig. 4k), indicating that taurocholate and taurodeoxycholate repress T3SS-1 gene expression via YedQ. Taken together, our data reveal that bile components taurocholate and taurodeoxycholate stimulate the DGC activity of YedQ to repress T3SS-1 gene expression in S. Typhimurium.
C-di-GMP affects expression and secretion of T3SS-1 effectors through binding to the T3SS-1 chaperone SicA
Based on the above observations, we speculated that there may exist a yet unidentified c-di-GMP-binding effector that regulates T3SS-1 gene expression at the transcriptional level. It was previously shown that the transcription factor InvF in complex with the T3SS-1 chaperone SicA directly activates the expression of SipB and SopB35,42. However, the mRNA levels of invF as well as its promoter activity were similar among the wild type, mutants ΔluxS and ΔyeaJ and the corresponding complemented strains (Supplementary Fig. 13), eliminating the possibility that an increase in invF expression results in elevated expression and production of SipB and SopB. A range of transcription factors have previously been shown to bind to c-di-GMP, leading to altered DNA binding capacity and thus changes in the expression of downstream target genes1,16. We thus examined whether InvF is a c-di-GMP effector that regulates downstream gene expression in response to this ligand. However, ITC analysis showed that InvF does not bind c-di-GMP (Supplementary Fig. 14a). Of note, a number of small proteins were reported to act as c-di-GMP-binding adaptors that regulate the catalytic or binding properties of their protein partners in a c-di-GMP-dependent manner20,21, thus raising another possibility that SicA is a c-di-GMP sensor. Intriguingly, binding analysis by ITC showed that SicA binds c-di-GMP at a 1:1 stoichiometry with a Kd of 0.21 ± 0.09 μM (Fig. 5a), which is comparable to previously reported Kd values for several well-established c-di-GMP receptors18-21. By contrast, no binding of c-di-AMP or cGMP to SicA was detected under the same experimental conditions (Supplementary Fig. 14b, c). These results indicate that c-di-GMP directly and specifically binds to SicA.
We further performed co-immunoprecipitation (co-IP) assays to investigate how c-di-GMP affects the interaction between SicA with InvF. We constructed two plasmids to express C-terminal His6-tagged InvF and hemagglutinin (HA)-tagged SicA, respectively, and found that the co-IP of InvF-His6 with SicA-HA was impaired by c-di-GMP in a dose-dependent fashion, but not by high concentrations of c-di-AMP or cGMP (Fig. 5b). We also performed electrophoretic mobility shift assays (EMSAs) to further evaluate the effect of c-di-GMP on the interactions of the InvF/SicA complex with its target promoters. The EMSA results showed that the InvF/SicA complex specifically binds to the promoter sequences of sopB, sopE2 and the sicAsipBCDA operon (Supplementary Fig. 15a-c). The addition of increasing concentrations of c-di-GMP reduced or even abrogated the formation of InvF/SicA-DNA complexes, whereas inclusion of c-di-AMP or cGMP at a dose corresponding to the highest level of c-di-GMP did not result in an observable difference in the InvF/SicA-DNA binding (Fig. 5c-e). Collectively, these data suggest that the binding of c-di-GMP to SicA inhibits the formation of the InvF/SicA complex, thus resulting in reduced binding of the complex to its target promoters.
SipB and SipC are T3SS-1 translocators that also act as effector proteins35,43,44. In addition to acting as a co-activator of InvF, SicA also functions to partition and stabilize SipB and SipC by binding directly to them45. Immunoprecipitation experiments revealed that SicA-HA co-precipitated both SipB-His6 and SipC-His6, while c-di-GMP decreased the co-IP of SipB-His6 and SipC-His6 with SicA-HA in a dose-dependent fashion (Fig. 5f, g). We thus hypothesized that c-di-GMP could also affect the stability and secretion of SipB and SipC at the post-translational level by inhibiting the binding of SicA to SipB and SipC. To experimentally test this hypothesis, we replaced the promoter of the sicAsipBCDA operon in the wild-type and mutant strains with the invF promoter whose activity is not regulated by c-di-GMP signaling (Supplementary Fig. 13). After promoter replacement, the expression of sopB and sopE2 with their own promoters was still significantly upregulated in ΔluxS and ΔyeaJ but dramatically reduced in ΔsicA when compared with the wild type, whereas the expression of sipB and sipC under the control of the invF promoter was not affected by deletion of luxS, yeaJ or sicA (Supplementary Fig. 16). In contrast, western blot analysis showed that intracellular accumulation and secretion of SipB and SipC were increased in ΔluxS and ΔyeaJ compared to the wild type, but reduced to very low levels in ΔsicA (Fig. 5h). These results support that changes in c-di-GMP concentration also play a role in the stability and secretion of SipB and SipC through targeting their chaperone SicA.
To study how SicA interacts with c-di-GMP, we constructed a homology model of SicA based on IpgC from Shigella flexneri (PDB ID: 3GYZ) using the Phyre2 server46. Potential ligand-binding sites were predicted using POCASA 1.147 and the molecular docking was performed by AutoDock Vina 1.1.241. The conformation with the lowest binding energy of −8.1 kcal mol−1 (Fig. 5i) suggests that c-di-GMP makes close contact with T25, K27, D28, Q34, D67, Y69, N70, P71 and D72 of SicA (Fig. 5j). Mutation of K27, D28, Q34, D67 or N70 resulted in a marked reduction in the c-di-GMP binding affinity for SicA (Fig. 5k and Supplementary Fig. 17), indicating that these residues are directly involved in c-di-GMP binding. In addition, protein-protein docking analysis by Cluspro 2.048 suggested that InvF, SipB and SipC have partially overlapping interaction surfaces on SicA, while the interaction surfaces of SicA with SipB and SipC, but not with InvF, partially overlap with the c-di-GMP-binding site (Supplementary Fig. 18 and Fig. 5j). Among residues of SicA that make contact to c-di-GMP, K27, D28, Q34 and D67, but not N70, were predicted to participate in interactions with its protein partners SipB and SipC (Supplementary Fig. 18). Indeed, the K27A variant showed a 19-fold lower binding affinity to SipB compared with wild-type SicA (Supplementary Fig. 19a, b). In contrast, the N70A mutation of SicA did not affect its binding affinities for InvF, SipB and SipC (Supplementary Fig. 19a, c-g). Moreover, changing N70 to alanine did not affect its ability to co-immunoprecipitate InvF-His6, SipB-His6 and SipC-His6 without addition of c-di-GMP, whereas high concentrations of c-di-GMP failed to impair co-IP of InvF-His6, SipB-His6 and SipC-His6 with SicAN70A-HA (Supplementary Fig. 20). These results indicate that changing N70 to alanine specifically impairs binding of c-di-GMP but leaves its chaperone function unaffected.
When SicAN70A was expressed at a level similar to that of wild-type SicA, the amounts of InvF, SipB and SipC bound by SicAN70A were much higher than those bound by wild-type SicA after the addition of the same concentrations of c-di-GMP (Supplementary Fig. 20), which can be attributed to the much lower c-di-GMP-binding affinity of SicAN70A and thus less binding of SicAN70A to c-di-GMP when compared with wild-type SicA. To determine the role of c-di-GMP on SicA activity in vivo, we replaced the wild-type sicA gene in the chromosome with sicA(N70A). While deletion of sicA significantly reduced the expression of sipB, sopB and sopE2, the expression levels of these genes were drastically increased in the sicA(N70A) mutant compared to the wild-type strain (Fig. 5l), indicating that less binding of SicAN70A to c-di-GMP but more binding of this chaperone to InvF leads to enhanced transcription of the target genes of InvF/SicA in the sicA(N70A) mutant. Consistent with previous findings that sicA is also a target gene of InvF/SicA35,42, the expression level of the sicA(N70A) gene in the sicA(N70A) mutant was significantly higher than that of sicA in the wild type (Fig. 5l). Furthermore, deletion of luxS or yeaJ in the sicA(N70A) mutant background did not alter expression of the T3SS-1 genes (Fig. 5l), suggesting that SicAN70A is unable to bind c-di-GMP at its physiological concentration. In the murine oral infection model, in contrast to the mutants ΔluxS and ΔyeaJ, the ΔsicA mutant led to significantly decreased mouse mortality compared to the wild-type strain (Fig. 5m). Complementation with plasmid-borne sicA restored the lethality of the ΔsicA mutant in mice to wild-type levels (Fig. 5m). By contrast, mice infected with the sicA(N70A) mutant showed significantly higher mortality than those infected with the wild type, whereas infections with sicA(N70A) and its derivative mutants lacking luxS or yeaJ produced similar mortality (Fig. 5m). These in vivo observations indicate that the N70A mutation of the sicA gene in S. Typhimurium promotes the T3SS-1 activity but abolishes responses to c-di-GMP signaling.
Taken together, these in vitro and in vivo results suggest that c-di-GMP exerts its regulatory effects on T3SS-1 through binding to SicA, and elevated intracellular levels of c-di-GMP will lead to less binding of SicA to InvF, SipB and SipC, thus downregulating expression of the T3SS-1 genes as well as impairing the stability and secretion of SipB and SipC.
c-di-GMP-binding SicA homologs are widely distributed among Gram-negative pathogenic bacteria
SicA of S. Typhimurium belongs to class II of T3SS chaperones and harbors 3 tandem tetratricopeptide repeat (TPR) motifs, which is characteristic for the CesD/SycD/LcrH family of T3SS chaperones49-51. Homology searches using the BLASTP program against the NCBI non-redundant protein database revealed that the CesD/SycD/LcrH family of chaperones that shares >20% sequence identity with SicA (E-value cutoff of 1E-03) are widely distributed in Gram-negative bacteria, especially the phylum Proteobacteria (Supplementary Data 2 and Supplementary Fig. 21). To examine whether c-di-GMP binding is a common feature of this family, T3SS chaperone proteins from several well-known pathogenic bacteria, including PcrH of Pseudomonas aeruginosa, IpgC of S. flexneri serotype 2a, SycD of Yersinia enterocolitica, CesD of EHEC O157:H7, BicA of Burkholderia thailandensis and VcrH of Vibrio parahaemolyticus, were expressed and purified as recombinant proteins from E. coli BL21(DE3) and their ability to bind c-di-GMP was assessed. Strikingly, while these 6 chaperones share 26.2-60.1% identity with SicA, all of them were found to bind c-di-GMP with high affinity (Kd = 0.15-0.22 μM) (Supplementary Fig. 22). Thus, our results suggest that the CesD/SycD/LcrH family of T3SS chaperones constitutes a large group of c-di-GMP effectors in Gram-negative pathogenic bacteria. Given the crucial roles of the CesD/SycD/LcrH family of chaperones in the synthesis and/or secretion of T3SS effectors50-52, our findings support the idea that the activity of T3SSs in a broad range of bacterial pathogens can be modulated by c-di-GMP signaling via this family of chaperones.