ABSTRACT
One important event for the divergence of Salmonella from Escherichia coli was the acquisition by horizontal transfer of the Salmonella pathogenicity island 1 (SPI-1), containing genes required for the invasion of host cells by Salmonella. HilD is an AraC-like transcriptional regulator in SPI-1 that induces the expression of the SPI-1 and many other acquired virulence genes located in other genomic regions of Salmonella. Additionally, HilD has been shown to positively control the expression of some ancestral genes (also present in E. coli and other bacteria), including phoH. In this study, we determined that both the gain of HilD and cis-regulatory evolution led to the integration of the phoH gene into the HilD regulon. Our results indicate that a HilD-binding sequence was generated in the regulatory region of the S. enterica serovar Typhimurium phoH gene, which mediates the activation of promoter 1 of this gene under SPI-1-inducing conditions. Furthermore, we found that repression by H-NS, a histone-like protein, was also adapted on the S. Typhimurium phoH gene and that HilD activates the expression of this gene in part by antagonizing H-NS. Additionally, our results revealed that the expression of the S. Typhmurium phoH gene is also activated in response to low phosphate but independently of the PhoB/R two-component system, known to regulate the E. coli phoH gene in response to low phosphate. Thus, our results indicate that cis-regulatory evolution has played a role in the expansion of the HilD regulon and illustrate the phenomenon of differential regulation of ortholog genes.
IMPORTANCE Two mechanisms mediating differentiation of bacteria are well known: acquisition of genes by horizontal transfer events and mutations in coding DNA sequences. In this study, we found that the phoH ancestral gene is differentially regulated between Salmonella Typhimurium and Escherichia coli, two closely related bacterial species. Our results indicate that this differential regulation was generated by mutations in the regulatory sequence of the S. Typhimurium phoH gene and by the acquisition by S. Typhimurium of foreign DNA encoding the transcriptional regulator HilD. Thus, our results, together with those from an increasing number of studies, indicate that cis-regulatory evolution can lead to the rewiring and reprogramming of transcriptional regulation, which also plays an important role in the divergence of bacteria through time.
KEYWORDS: AraC-like, cis-regulatory, Escherichia coli, H-NS, HilD, PhoB/R, regulatory evolution, SPI-1, Salmonella, phoH
INTRODUCTION
The acquisition of foreign DNA through horizontal transfer events is one of the main genetic processes that influences the dynamics of bacterial genomes (1). Likewise, the adaptation of regulatory mechanisms to control the expression of acquired genes, or even to modify the expression pattern of ancestral genes, has been pivotal for evolution of bacteria, including pathogenic species (2–5). During its divergence from Escherichia coli, about 100 million years ago, the Gram-negative pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) shaped approximately a quarter of its genome with acquired DNA (6). Most of the genes for S. Typhimurium virulence have been acquired by horizontal transfer events and are located in discrete regions of the genome called Salmonella pathogenicity islands (SPI), which are not present in E. coli (3, 6). SPI-1, a cluster of 39 genes, is essential for the S. Typhimurium invasion of intestinal epithelium cells, which leads to enteritis (7, 8).
HilD is a member of the AraC/XylS family of transcriptional regulators, located in SPI-1, which directly or through distinct regulators positively controls the expression of the SPI-1 genes and several other related genes located outside this island (2, 3, 9–13). Interestingly, HilD has been shown to induce not only the expression of numerous acquired genes but also that of some ancestral genes (those present in E. coli and other phylogenetically distant bacteria). For instance, HilD positively regulates the expression of the flhDC ancestral operon, which encodes the master complex regulator of flagellar and chemotaxis genes, FlhD4C2 (14, 15). Furthermore, HilD induces the expression of the yobH ancestral gene that codes for the 79-amino-acid polypeptide YobH (11, 12, 16). Importantly, both the flagellar/chemotaxis genes and the yobH gene are necessary for the Salmonella invasion of host cells (16–20). Therefore, HilD has played a major role in the evolution of Salmonella pathogenicity by integrating the expression of tens of acquired genes and some ancestral genes into a big virulence regulon.
Previously, we and others found that HilD also induces the expression of the phoH ancestral gene under SPI-1-inducing conditions, such as the growth of bacteria in nutrient-rich lysogeny broth (LB) (10–12). Orthologs of the PhoH protein are present in E. coli and many other bacteria as well as in some archaea; however, the function of PhoH remains unknown in all microorganisms (21, 22). In E. coli, it has been demonstrated that the expression of phoH is induced by the PhoB/R two-component system in minimal medium containing a low concentration of phosphate; additionally, it was found that PhoH shows ATPase activity (23). The PhoB/R system positively regulates the expression of several other genes in response to low phosphate, including the pstS gene; the response regulator PhoB binds to a sequence denominated pho-box that is present on the regulatory regions of its target genes (24).
Remarkably, to our knowledge, in addition to the PhoB/R system (or orthologs), HilD is the only other regulator that has been involved in the expression of phoH. Hence, in this study we aimed to uncover the elements that led to the integration of the phoH gene into the HilD regulon.
By comparing the expression of the phoH gene from S. Typhimurium and E. coli, we show that the gain of HilD and cis-regulatory evolution led to the regulation of phoH by HilD in S. Typhimurium. Furthermore, our results support that HilD activates the expression of phoH under SPI-1-inducing conditions in part by acting as an anti-H-NS factor. Additionally, our results revealed differential regulation of the phoH gene in response to low phosphate; in E. coli this regulation is mediated by the PhoB/R system, whereas in S. Typhimurium it is independent of PhoB.
RESULTS
The S. Typhimurium phoH gene, but not that of E. coli K-12, is expressed and regulated by HilD under SPI-1-inducing conditions.
The phoH structural gene and its encoded protein, PhoH, share identities of 82% and 95%, respectively, between S. Typhimurium SL1344 and E. coli MG1655. In contrast, the intergenic sequences upstream of phoH show a lower identity (54%), and the genomic context is poorly conserved between these two bacteria (Fig. 1A). Therefore, we wondered whether, in addition to the gain of HilD by S. Typhimurium, differentiation of the phoH regulatory sequence in S. Typhimurium was also necessary for the adaptation of the regulation of phoH by HilD. To examine this idea, we analyzed the expression of the phoH gene from S. Typhimurium SL1344 (phoHSTm) and from E. coli MG1655 (phoHEc) in S. Typhimurium and E. coli strains expressing HilD or not and grown under SPI-1-inducing conditions (LB at 37°C). First, we constructed the phoHEc-cat transcriptional fusion (carrying the full-length intergenic sequence upstream of phoHEc), which is equivalent to the phoHSTm-cat transcriptional fusion that we previously used to show the regulation of the phoHSTm gene by HilD (10) (Fig. 1B, Table 1). The expression of both fusions, phoHSTm-cat and phoHEc-cat, then was quantified in the wild-type (WT) S. Typhimurium SL1344 strain and its derivative ΔhilD mutant, as well as in the WT E. coli MC4100 strain, in the presence of the pK6-HilD plasmid expressing HilD or the pMPM-K6 vector. Consistent with our previous study (10), the expression of the phoHSTm-cat fusion was significantly decreased in the ΔhilD+vector mutant and in the WT E. coli+vector strain with respect to the WT S. Typimurium+vector strain; furthermore, it was induced by the pK6-HilD plasmid in both the ΔhilD mutant and the WT E. coli strain, 4- and 8-fold, respectively (Fig. 2A). In contrast, the phoHEc-cat fusion showed a low expression level in all the strains tested (Fig. 2B), indicating that the expression of the phoHEc gene is not activated by HilD under the growth conditions tested. To confirm these results, we performed a Western blot analysis to monitor the chromosomal expression of the PhoH-FLAG protein (PhoH tagged with a 3×FLAG epitope) in the S. Typhimurium ΔhilD mutant and the WT E. coli strain carrying the pBAD-HilD1 plasmid, which expresses HilD from an arabinose-inducible promoter. In agreement with our results obtained with the transcriptional fusions, the S. Typhimurium PhoH-FLAG (PhoHStm-FLAG), but not the E. coli PhoH-FLAG (PhoHEc-FLAG), was detected under the conditions assessed; furthermore, the expression of HilD from the pBAD-HilD1 plasmid increased the amount of PhoHStm-FLAG in the ΔhilD mutant in a dose-dependent fashion (Fig. 2C). These results show that the phoHSTm gene, but not the phoHEc gene, is expressed and regulated by HilD under SPI-1-inducing conditions, which supports that both the gain of HilD and differentiation of the regulatory sequence led the phoHSTm gene to be expressed under conditions relevant for Salmonella virulence.
FIG 1.
Genomic context of the phoHSTm and phoHEc genes and schematic representation of the phoHSTm-cat and phoHEc-cat fusions. (A) Genomic context of the phoH gene from the S. Typhimurium SL1344 and E. coli K-12 MG1655 strains. Genes are shown by arrows. Homologous genes are depicted with the same color. (B) Schematic representation of the phoHSTm-cat and phoHEc-cat transcriptional fusions containing the full-length upstream region of phoH from S. Typhimurium SL1344 or E. coli K-12 MG1655 strains, respectively. Transcriptional start sites are shown by bent arrows. Positions indicated are relative to the transcriptional start site of promoter 1 of each gene.
TABLE 1.
Bacterial strains and plasmids used in this studya
| Strain or plasmid | Genotype or description | Reference or source |
|---|---|---|
| Strains | ||
| S. Typhimurium | ||
| SL1344 | Wild type; xyl, hisG, rpsL; Smr | 60 |
| DTM142 | phoH::3×FLAG-kan | This study |
| JPTM25 | ΔhilD | 47 |
| DTM143 | ΔhilD phoH::3×FLAG | This study |
| DTM144 | ΔphoB::kan | This study |
| DTM145 | ΔphoB | This study |
| DTM146 | ΔphoB phoH::3×FLAG-kan | This study |
| DTM150 | ΔphoH::kan | This study |
| DTM151 | ΔsiiE::kan | This study |
| E. coli | ||
| MG1655 | Wild type; prototrophic E. coli K-12 | Michael Cashel |
| DH10β | Laboratory strain | Invitrogen |
| MC4100 | F− (araD139) Δ(argF-lac)169λ− e14− flhD5301 Δ(fruK-yeiR) 725(fruA25) relA1 rpsL150(SmR) rbsR22 Δ(fimB-fimE)632(::IS1)deoC1 | 61 |
| BL21/DE3 | Strain for expression of recombinant proteins | Invitrogen |
| JPMC34 | MC4100 Δhns | 62 |
| JW0389 | BW25113 ΔphoB::kan | 63 |
| DTM147 | phoH::3×FLAG-kan | This study |
| DTM148 | BW25113 ΔphoB | This study |
| DTM149 | BW25113 ΔphoB phoH::3×FLAG-kan | This study |
| Plasmids | ||
| pKK232-8 | pBR322 derivative containing a promoterless chloramphenicol acetyltransferase (cat) gene, Apr | 48 |
| pphoHSTm-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of S. Typhimurium from nucleotides −653 to +416 | 10 |
| pphoHEc-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of E. coli from nucleotides −406 to +406 | This study |
| pP1phoHSTm-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of S. Typhimurium from nucleotides −653 to +12 | This study |
| pP2phoHSTm-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of S. Typhimurium from nucleotides −5 to +416 | This study |
| pP1phoHSTm-248 + 12-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of S. Typhimurium from nucleotides −248 to +12 | This study |
| pP1phoHSTm−67 + 3-cat | pKK232-8 derivative containing a phoH-cat transcriptional fusion of S. Typhimurium from nucleotides −67 to +3 | This study |
| pphoHHilDmut-cat | pphoHSTm-cat derivative containing mutations in the predicted HilD-binding site | This study |
| pphoHP1mut-cat | pphoHSTm-cat derivative containing mutations in the P1 promoter | This study |
| ppstSSTm-cat | pKK232-8 derivative containing a pstS-cat transcriptional fusion of S. Typhimurium from nucleotides −232 to +352 | This study |
| pBADMycHisC | Expression vector for constructing C-terminal MycHis fusions under an arabinose-inducible promoter, Apr | Invitrogen |
| pBAD-HilD1 | pBADMycHis derivative expressing HilD-MycHis under an arabinose-inducible promoter | 47 |
| pMPM-K6Ω | p15A derivative cloning vector containing an arabinose-inducible promoter, Kanr | 64 |
| pK6-HilD | pMPM-K6Ω derivative expressing HilD under an arabinose-inducible promoter, Kanr | 10 |
| pMAL-HilD1 | pMAL-c2X derivative expressing MBP-HilD from a lac promoter, Apr | 2 |
| pBAD-H-NS-FH | pBADMycHisC derivative expressing H-NS-FH from an arabinose-inducible promoter, Apr | 57 |
| pKD46 | pINT-ts derivative expressing red recombinase under an arabinose-inducible promoter, Apr | 49 |
| pKD4 | pANTsγ derivative template plasmid containing the kanamycin cassette for λRed recombination, Apr | 49 |
| pSUB11 | pGP704 derivative template plasmid for FLAG epitope tagging | 50 |
| pFLP3 | pFLP2 derivative template plasmid expressing yeast Flp recombinase, Apr, Tcr | 51 |
The coordinates for the cat fusions are indicated with respect to the transcriptional start site of pstS or that of the P1 promoter of phoHSTm or phoHEc. Apr, ampicillin resistance; Smr, streptomycin resistance; Kanr, kanamycin resistance; Tcr, tetracycline resistance.
FIG 2.
phoHSTm, but not phoHEc, is expressed and regulated by HilD under SPI-1-inducing conditions. CAT-specific activity of the phoHSTm-cat (A) and phoHEc-cat (B) transcriptional fusions, contained in the pphoHSTm-cat and pphoHEc-cat plasmids, respectively, was determined in the WT S. Typhimurium (STm) SL1344 strain, its derivative ΔhilD mutant, and the WT E. coli (Ec) MC4100 strain, in the presence of the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD from an arabinose-inducible promoter. Expression of HilD from the pK6-HilD plasmid was induced with 0.001% l-arabinose added to the medium at the beginning of cultures. Data represent the averages with standard deviations from three independent experiments performed in duplicate. Statistically different values are indicated (*, P < 0.05; ****, P < 0.0001). (C) Expression of the PhoH-FLAG protein in the S. Typhimurium ΔhilD mutant and the WT E. coli (Ec) MG1655 strain carrying the vector pBADMycHisC (−) or the pBAD-HilD1 (+) plasmid, which expresses HilD from an arabinose-inducible promoter, was analyzed by Western blotting using monoclonal anti-FLAG antibodies. As a control, the expression of GroEL was also detected using polyclonal anti-GroEL antibodies. Expression of HilD from the pBAD-HilD1 plasmid was induced with the indicated amounts of l-arabinose added to the medium 2 h postinoculation of cultures. CAT activity and detection of the PhoH-FLAG and GroEL proteins were determined from samples of bacterial cultures grown in LB at 37°C for 9 and 4 h, respectively.
HilD specifically induces the expression of the P1 promoter of phoHSTm.
Previous transcriptomics studies revealed two transcription start sites (TSSs) for the phoHSTm gene (25, 26). To determine whether these TSSs are really generated by two different promoters (P1 and P2) and whether HilD regulates both or only one of them, we constructed and analyzed the expression of cat transcriptional fusions only containing the putative promoter P1 (P1phoHSTm-cat) or P2 (P2phoHSTm-cat) (Fig. 3A). The expression of these fusions was quantified in the WT S. Typhimurium strain and its derivative ΔhilD mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD under SPI-1-inducing conditions. The two fusions showed expression in the WT+vector strain, indicating that both P1 and P2 promoters are active under the conditions tested, with the P1 promoter 2-fold more active than the P2 promoter (Fig. 3B). However, only the expression of the P1phoHSTm-cat fusion decreased in the ΔhilD+vector mutant and was induced by the presence of the pK6-HilD plasmid (Fig. 3B). In agreement with these results, in electrophoretic mobility shift assays (EMSAs), purified MBP-HilD protein bound the phoHSTm fragment contained in the P1phoHSTm-cat fusion but not that contained in the P2phoHSTm-cat fusion (Fig. 3C). Together, these results support that HilD regulates only the P1 promoter of the phoHSTm gene.
FIG 3.
HilD directly regulates the P1 promoter of phoHSTm. (A) Schematic representation of the P1phoHSTm-cat and P2phoHSTm-cat transcriptional fusions containing promoter 1 (P1) or 2 (P2) of phoHSTm, respectively. Transcriptional start sites (bent arrows) and promoters (blue and red boxes) are shown. Positions indicated are relative to the transcriptional start site of promoter 1. (B) CAT-specific activity of the P1phoHSTm-cat and P2phoHSTm-cat transcriptional fusions, contained in the pP1phoHSTm-cat and pP2phoHSTm-cat plasmids, respectively, was determined in the WT S. Typhimurium (STm) SL1344 strain and its derivative ΔhilD mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD from an arabinose-inducible promoter. Expression of HilD from the pK6-HilD plasmid was induced with 0.001% l-arabinose added to the medium at the beginning of cultures. CAT activity was quantified from samples of bacterial cultures grown in LB at 37°C for 9 h. Data represent averages with standard deviations from three independent experiments performed in duplicate. Statistically different values are indicated (****, P < 0.0001). (C) MBP-HilD binding to the phoHSTm fragments contained in the P1phoHSTm-cat (P1) and P2phoHSTm-cat (P2) fusions was analyzed by competitive nonradioactive EMSAs. The DNA-protein complexes are indicated by an asterisk. Nonspecific PCR products are indicated by an arrow.
Cis-regulatory evolution led to the regulation of the P1 phoHSTm promoter by HilD.
To delimit the sequence required for the control of P1phoHSTm by HilD, we constructed and analyzed the expression of two additional cat fusions carrying different 5′ and 3′ deletions regarding the P1phoHSTm-cat-653 + 12 fusion used initially (Table 1). The expression of these two new fusions, P1phoHSTm-cat-248 + 12 and P1phoHSTm-cat-67 + 3, was similarly decreased in the ΔhilD+vector mutant with respect to the WT+vector strain and was induced in the ΔhilD mutant by the presence of the pK6-HilD plasmid (Fig. 4A). These results reveal that HilD acts on the sequence between positions −67 and +3 of the P1 phoHSTm promoter (P1phoHSTm), the sequence contained in the P1phoHSTm-cat-67 + 3 fusion. In agreement with this conclusion, a HilD-binding site was predicted between positions −59 and −25 of P1phoHSTm, which shows 62% A+T content (Fig. 4B). Previous studies indicate that HilD binding sites have a high A+T content (14, 27).
FIG 4.
HilD induces the expression of phoHSTm by acting on a regulatory sequence differentiated between Salmonella and E. coli. (A) CAT-specific activity of the P1phoHSTm-cat-248 + 12 and P1phoHSTm-cat−67 + 3 transcriptional fusions, contained in the pP1phoHSTm-248 + 12-cat and pP1phoHSTm−67 + 3-cat plasmids, respectively, was determined in the WT S. Typhimurium (STm) SL1344 strain and its derivative ΔhilD mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD from an arabinose-inducible promoter. Expression of HilD from the pK6-HilD plasmid was induced with 0.001% l-arabinose added to the medium at the beginning of cultures. CAT activity was quantified from samples of bacterial cultures grown in LB at 37°C for 9 h. Data represent averages with standard deviations from three independent experiments performed in duplicate. Statistically different values are indicated (****, P < 0.0001). (B) Alignment of the P1 −67/+3 sequences of S. Typhimurium (STm) SL1344 and E. coli (Ec) MG1655. The predicted HilD-binding site on the STm sequence and the PhoB-binding site (pho-box) on the E. coli sequence as well as the mutations generated in the predicted HilD-binding site and in the −10 region of the P1 phoHSTm promoter (HilDmut and P1mut, respectively) are shown. Bent arrows indicate the transcriptional start site of the respective P1 promoter. Asterisks indicate conserved nucleotides. (C) Phylogenetic tree based on the P1 −67/+3 sequence of E. coli O104:H4 (EAEC), E. coli O157:H7 (EHEC), E. coli K-12 MG1655, E. coli O78:H11 (ETEC), E. coli O127:H6 (EPEC), E. coli STEC_7v (STEC), E. coli K-12 ATCC 1175, E. coli 26-1 (UPEC), S. bongori, S. Choleraesuis SC-B67, S. Gallinarum 9184, S. Enteritidis 22510-1, S. Paratyphi A ATCC 11511, S. Typhi Ty2, S. Typhimurium SL1344, and S. Heidelberg 41563. Identity with respect to the sequence of S. Typhimurium SL1344 is shown with a heat map.
To confirm that HilD mediates expression of phoHSTm by acting on the P1 promoter of this gene, the predicted HilD-binding site or the −10 region of the P1 promoter were mutated, as indicated in Fig. 4B, in the phoHSTm-cat-653 + 416 fusion carrying the full-length regulatory region of phoHSTm (contains the P1 and P2 promoters), generating the phoHHilDmut-cat and phoHP1mut-cat fusions, respectively. The expression of the WT and its derivative mutated transcriptional fusions was quantified in the WT S. Typhimurium strain and the ΔhilD mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD under SPI-1-inducing conditions. As previously observed (Fig. 2A), the WT phoHSTm-cat fusion showed regulation by HilD, that is, its expression was decreased and increased in the ΔhilD+vector and ΔhilD+pK6-HilD strains, respectively (Fig. 5A). In contrast, both the phoHHilDmut-cat and phoHP1mut-cat fusions showed a low expression level that was similar in the three strains tested (Fig. 5A), which indicates that the regulation of phoHSTm by HilD is lost when the predicted HilD-binding site or the P1 promoter are mutated. EMSAs confirmed that HilD indeed binds the predicted site upstream of the P1 promoter (Fig. 5B to D). The MBP-HilD protein used in the EMSAs apparently forms overly large complexes that remained near the wells of the gel, with or without the tested DNA fragments; the WT P1phoH-157/+12 fragment and that carrying the mutated P1 promoter, but not that carrying the mutated HilD-binding sequence, were bound by MBP-HilD (Fig. 5B to D). Together, these results show that HilD positively and specifically regulates the P1 promoter of phoHSTm by binding a site that overlaps the −35 region of this promoter.
FIG 5.
HilD positively regulates phoHSTm by binding a site that overlaps the P1 promoter of this gene. (A) CAT-specific activity of the phoHSTm-cat, phoHHilDmut-cat, and phoHP1mut-cat transcriptional fusions, contained in the pphoHSTm-cat, pphoHHilDmut-cat, and pphoHP1mut-cat plasmids, respectively, was determined in the WT S. Typhimurium (STm) SL1344 strain and its derivative ΔhilD mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD from an arabinose-inducible promoter. Expression of HilD from the pK6-HilD plasmid was induced with 0.001% l-arabinose added to the medium at the beginning of cultures. CAT activity was quantified from samples of bacterial cultures grown in LB at 37°C for 9 h. Data represent the averages with standard deviations from three independent experiments performed in duplicate. Statistically different values are indicated (*, P = 0.0148; ****, P < 0.0001). MBP-HilD binding to the WT P1phoH-157/+12 fragment (B), the P1phoHHilDmut-157/+12 fragment mutated in the predicted HilD-binding site (C), and the P1phoHP1mut-157/+14 fragment mutated in the P1 promoter (D) was analyzed by nonradioactive EMSAs. The upper panels show the free DNA and the DNA-protein complexes stained with ethidium bromide, and the lower panels show the immunodetection of MBP-HilD at the top of the lines using anti-MBP monoclonal antibodies. The DNA-protein complexes are indicated by an asterisk.
Two promoters have also been reported for the phoHEc gene (23) that are located in positions similar to those of the phoHSTm gene (Fig. 1B). As shown in Fig. 2, the phoHEc gene is not regulated by HilD, which indicates that it lacks the HilD-acting site located upstream of P1phoHSTm. To investigate this notion, we performed an alignment of the corresponding −67/+3 sequence of the phoHSTm and phoHEc P1 promoters (P1 −67/+3 sequences). Interestingly, these sequences share only 45% identity; moreover, the nucleotides of the putative HilD-binding site located on the S. Typhimurium sequence are poorly conserved in the E. coli sequence (Fig. 4B). Consistent with this, a HilD-binding site was not predicted in the E. coli P1 −67/+3 sequence. To explore whether the differences shown by the P1 −67/+3 sequence of phoHSTm and phoHEc are a product of an evolutionary divergence between Salmonella and E. coli or are specific to the bacterial strains tested, we extended the analysis of the P1 −67/+3 sequence of phoH to other S. enterica serovars, the S. bongori species, and other K12 strains and pathotypes of E. coli. As shown in Fig. 4C, phylogenetic analysis based on the P1 −67/+3 sequence of phoH grouped the Salmonella and E. coli strains tested in two different clades. In agreement with these results, the S. Typhimurium P1 −67/+3 sequence is better conserved in the S. enterica serovars (94 to 100% identity), and to a lower extent in S. bongori (73% identity), than in the E. coli strains assessed (54 to 56% identity) (Fig. 4C). These results suggest that evolution led to the differentiation of the P1 −67/+3 sequence between Salmonella and E. coli.
Together, our results support that the regulation of the phoHSTm gene by HilD was adapted by the gain of HilD and the generation of a HilD-acting site through cis-regulatory evolution.
phoHSTm and phoHEc are differentially affected by the H-NS repressor.
HilD induces the expression of target genes by counteracting H-NS-mediated repression; in the absence of H-NS activity, these genes are expressed independently of HilD (2, 3, 16, 27–30). We sought to determine whether HilD follows a similar mechanism to induce the expression of the phoHSTm gene. Salmonella hns mutants show severe growth defects due to the overexpression of SPI-1 and other related genes, and suppressor mutations are generated (31, 32). Thus, to analyze the effect of H-NS on the phoHSTm gene, we used the WT E. coli MC4100 strain and its derivative Δhns mutant that does not show growth defects; the E. coli genetic background has been useful to study the regulation of other Salmonella genes by H-NS (H-NS of S. Typhimurium SL1344 and E. coli MC4100 share 95% identity) (16, 30). On the other hand, HilD induces the expression of phoHSTm in E. coli K-12 (Fig. 2A), indicating that no other Salmonella-specific regulator is required for the HilD-mediated expression of this gene. We quantified the expression of the phoHSTm-cat fusion in the WT E. coli MC4100 strain and its derivative Δhns mutant carrying the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD, grown under SPI-1-inducing conditions. As a control, the expression of this fusion was also tested in the WT S. Typhimurium strain carrying the pMPM-K6 vector. In line with the results described above (Fig. 2A), the expression of the phoHSTm-cat fusion was induced in the WT S. Typhimurium+vector strain as well as in the WT E. coli+pK6-HilD strain but not in the E. coli+vector strain (Fig. 6A). Notably, in the E. coli Δhns+vector strain the expression of the phoHSTm-cat fusion reached levels similar to those in the WT S. Typhimurium strain (Fig. 6A). These results show that H-NS represses the expression of phoHSTm and that in the absence of H-NS this gene is expressed independently of HilD, which supports that HilD induces expression of phoHSTm by counteracting H-NS-mediated repression. Nevertheless, the presence of the pK6-HilD plasmid further increased (2-fold) the expression of the phoHSTm-cat fusion in the E. coli Δhns mutant (Fig. 6A), indicating that HilD also induces partial expression of the phoHSTm gene by a mechanism independent of H-NS.
FIG 6.
HilD antagonizes H-NS-mediated repression on phoHSTm. (A) CAT-specific activity of the phoHSTm-cat transcriptional fusion, contained in the pphoHSTm-cat plasmid, was evaluated in the WT S. Typhimurium (STm) SL1344 strain as well as in the WT E. coli (Ec) MC4100 strain and its derivative Δhns mutant in the presence of the pMPM-K6 vector or the pK6-HilD plasmid expressing HilD from an arabinose-inducible promoter. CAT-specific activity of the phoHEc-cat transcriptional fusion, contained in the pphoHEc-cat plasmid, was evaluated in the WT E. coli (Ec) MC4100 strain and its derivative Δhns mutant. Expression of HilD from the pK6-HilD plasmid was induced with 0.001% l-arabinose added to the medium at the beginning of cultures. CAT activity was quantified from samples of bacterial cultures grown in LB at 37°C for 9 h. Data represent the average with standard deviations from three independent experiments performed in duplicate. Statistically different values are indicated (****, P < 0.0001). (B) H-NS-FH binding to the phoH fragments contained in the phoHSTm-cat and phoHEc-cat fusions, carrying the full-length regulatory region of phoHSTm and phoHEc, respectively, was analyzed by competitive nonradioactive EMSAs. The DNA-protein complexes are indicated by an asterisk. Nonspecific PCR products are indicated by an arrow. (C) Competitive nonradioactive EMSAs between HilD-MBP and H-NS-FH on the P1phoH-157/+12 fragment of phoHSTm. Purified H-NS-FH protein was added at 4 μM (lanes 3 to 6), and purified MBP-HilD protein was added at 0.4, 0.8, and 1.2 μM (lanes 4 to 6, respectively). No protein was added in lane 1, and MBP-HilD was added at 1.2 μM in lane 2. In EMSAs, the upper panels show the free DNA and the DNA-protein complexes stained with ethidium bromide, whereas the lower panels show the immunodetection of H-NS-FH in the DNA-protein complexes at the top (B) or within the lanes of the gels (C), using anti-FLAG monoclonal antibodies.
We next assessed whether H-NS also represses the expression of the phoHEc gene. For this, the expression of the phoHEc-cat fusion was quantified in the WT E. coli and E. coli Δhns strains grown under SPI-1-inducing conditions. The phoHEc-cat fusion showed very low expression levels in both E. coli strains (Fig. 6A), showing that, in contrast to that observed for phoHSTm, the expression of phoHEc is not induced by the absence of H-NS under the conditions tested.
EMSAs were performed to determine whether H-NS regulates phoHSTm directly. As could be expected, purified H-NS-FH (H-NS-Flag-His6) protein bound the DNA fragment spanning the entire regulatory region of phoHSTm, but not that of phoHEc, at the concentrations tested (Fig. 6B). The DNA-H-NS-FH complexes formed with the entire regulatory region of phoHSTm seem to be very large, since they were retained near the wells of the gel (Fig. 6B); with a shorter fragment of the regulatory region of phoHSTm, the DNA-H-NS-FH complexes were detected within the gel (Fig. 6C). H-NS is known to bind A+T-rich sequences (33); intriguingly, both the regulatory region of phoHSTm and that of phoHEc show high A+T content, 57% and 55%, respectively. Next, we analyzed by competitive EMSAs whether MBP-HilD can displace H-NS-FH from phoHSTm. Binding reactions containing the P1phoH-157/+12 fragment of phoHSTm were first incubated with a constant concentration of H-NS-FH, and then increasing concentrations of MBP-HilD were added. Binding reactions containing only H-NS-FH or MBP-HilD were also tested. The DNA-H-NS-FH complex was shifted by MBP-HilD to a slower-migrating complex that remained near the wells of the gel, similar to the complex formed only by MBP-HilD; furthermore, MBP-HilD decreased the amount of H-NS-FH bound to the P1phoH-157/+12 fragment (Fig. 6C). These results indicate that HilD displaces H-NS from phoHSTm.
In all, these results support that cis-regulatory evolution led to the repression of phoHSTm by H-NS, which is involved in the regulation of this gene by HilD.
phoHSTm and phoHEc are differentially regulated in response to inorganic phosphorus.
The phoHEc gene belongs to the phosphate regulon; its expression is induced by the PhoR/B two-component system in response to inorganic phosphorus (Pi) starvation (22, 23). It was therefore of interest to test whether the phoHSTm gene that was recruited into the HilD regulon conserves the ancestral regulation by the PhoR/B system in response to low Pi. To investigate this notion, we quantified the expression of the phoHSTm-cat fusion in the WT S. Typhimurium strain and its isogenic ΔphoB mutant grown in N-minimal medium (N-MM) containing low or high concentrations of Pi. Surprisingly, the expression of the phoHSTm-cat fusion was induced by low Pi in both the WT strain and the ΔphoB mutant (Fig. 7A). Similar results were obtained by analyzing the expression of the PhoHStm-FLAG protein (Fig. 7B). In contrast, the expression of the PhoHEc-FLAG protein or the phoHEc-cat fusion was induced by low Pi in the WT E. coli strain but not, or only slightly, in its derivative ΔphoB mutant (Fig. 7C and D). In agreement with these results, an in silico analysis identified the previously reported pho-box (PhoB-binding site) on the regulatory sequence of phoHEc but did not predict a pho-box on the corresponding sequence of phoHSTm (Fig. 4B) (23). The S. Typhimurium sequence conserves only 10 of the 18 nucleotides forming the E. coli pho-box (Fig. 4B). These results indicate that both phoHEc and phoHSTm are similarly regulated by Pi but through different mechanisms; the one for phoHEc involves PhoB and that for phoHSTm does not.
FIG 7.
Expression of phoHSTm is activated by low Pi independently of either PhoB or HilD. CAT-specific activity of the phoHSTm-cat (A) and pstSSTm-cat (E) transcriptional fusions, contained in the pphoHSTm-cat and ppstSSTm-cat plasmids, respectively, was determined in the WT S. Typhimurium (STm) SL1344 strain and its derivative ΔphoB and ΔhilD mutants. pstS is known to be regulated by PhoB; thus, it was tested as a positive control. (C) CAT-specific activity of the phoHEc-cat transcriptional fusion, contained in the pphoHEc-cat plasmid, was determined in the WT E. coli (Ec) BW25113 strain and its derivative mutant. Data represent the average with standard deviation from three independent experiments performed in duplicate. Statistically different values are indicated (*, P < 0.0108; ****, P < 0.0001). The expression of the PhoHSTm-FLAG protein in the WT S. Typhimurium (STm) SL1344 strain and its derivative ΔphoB and ΔhilD mutants (B), as well as the expression of the PhoHEc-FLAG protein in the WT E. coli (Ec) MG1655 strain and its derivative ΔphoB mutant (D), was analyzed by Western blotting using monoclonal anti-FLAG antibodies. As a control, the expression of GroEL was also detected using polyclonal anti-GroEL antibodies. CAT activity and detection of the PhoH-FLAG and GroEL proteins were determined from samples of bacterial cultures grown for 16 h at 37°C in N-MM containing low (L) or high (H) Pi.
Next, we asked whether the regulation by Pi independent of PhoB also acts on the pstS gene, which has been shown to belong to the phosphate regulon and to be regulated by PhoB in different bacteria; this gene encodes a component of a high-efficiency Pi transporter (24). To know this, a cat transcriptional fusion of the S. Typhimurium SL1344 pstS (pstSSTm) gene was constructed and assessed in the same bacterial strains and growth conditions as in the previous experiment. The expression of the pstSSTm-cat fusion was induced by low Pi in the WT S. Typhimurium strain but not in its derivative ΔphoB mutant (Fig. 7E), indicating that the pstSSTm gene is regulated by Pi starvation through PhoB. Consistent with this, a computational analysis identified on pstSSTm (data not shown) the pho-box previously reported for the E. coli gene (34).
As expected, according to different studies indicating that HilD does not affect expression of target genes when S. Typhimurium is grown in minimal medium (2, 25), the regulation of phoHSTm and pstSSTm by Pi was similar in the WT strain and its derivative ΔhilD mutant (Fig. 7A, B, and E).
Taken together, these results support that the phoHSTm gene evolved to be regulated by Pi through a PhoB-independent mechanism.
DISCUSSION
Differential regulation of homologous genes is an important mechanism for phenotypic variability among closely related bacteria (4, 35–38). In this study, we further define the mechanism by which HilD induces the expression of phoHSTm and show that both the gain of HilD and cis-regulatory evolution adapted this regulation in S. Typhimurium, leading to differential expression of phoH between S. Typhimurium and E. coli K-12.
Our data indicate that in the absence of the H-NS repressor, the phoHSTm gene reached WT expression levels independently of HilD. We found that H-NS binds to and represses the expression of the phoHSTm gene but not the phoHEc gene. Moreover, we determined that HilD displaces H-NS from the phoHSTm gene. These results support that HilD induces the expression of phoHSTm by antagonizing the negative effect of H-NS, a mechanism that HilD follows to induce the expression of several other target genes (2, 9, 16, 27–29, 39). Interestingly, we observed that the overexpression of HilD further increased the expression of the phoHSTm gene in the absence of H-NS, which suggests that HilD, besides acting as an anti-H-NS factor, could antagonize the action of an additional repressor or improve the RNA polymerase binding on the promoter of this gene. In agreement with this notion, a previous report indicates that HilD counteracts repression exerted by the nucleoid-associated protein Hha on the rtsA gene (27). Furthermore, evidence from another study suggests that HilD recruits the RNA polymerase on the hilA promoter (40).
A previous transcriptome sequencing (RNA-seq) analysis revealed two transcriptional start sites for the phoHSTm gene (25, 26). Accordingly, our results show that two promoters (P1 and P2) transcribe this gene under SPI-1-inducing conditions. We found that HilD positively regulates the P1 promoter, but not the P2 promoter, by binding a site that overlaps the −35 region of the P1 promoter. To our knowledge, not one regulatory mechanism other than that mediated by the PhoB/R system (see below) had been defined for the phoH gene in any other bacteria.
We detected only background expression levels for the phoHEc gene under SPI-1-inducing conditions, even in the presence of HilD. In agreement with this finding, computational analyses revealed that the regulatory sequence required for the HilD-mediated expression of the phoHSTm gene (P1 −67/+3 sequence) is poorly conserved in the phoHEc gene, including the HilD-binding sequence on phoHSTm. Moreover, a phylogenetic analysis based on the P1 −67/+3 sequence of phoH grouped Salmonella and E. coli strains in two different clades. Thus, our results support that both the acquisition of HilD and regulatory evolution of the P1 −67 to +3 sequence adapted the regulation of phoH by HilD in S. Typhimurium, which led to this gene being expressed under growth conditions relevant for Salmonella virulence. Interestingly, cis-regulatory evolution also adapted repression of phoHSTm by H-NS, which is involved in the HilD-mediated expression of this gene. It is tempting to speculate that the regulation by HilD of other ancestral genes, like flhDC and yobH, was also adapted by cis-regulatory evolution. On the other hand, cis-regulatory changes in S. Typhimurium with respect to S. bongori led the srfN, ugtL, and flhDC genes to be regulated by the SPI-2 transcriptional regulator SsrB and thus to have a role in S. Typhimurium virulence (36–38). Thus, these reports indicate that cis-regulatory changes have played an important role in the evolution of Salmonella pathogenicity. It is reasonable to think that the integration of the phoHSTm gene into the HilD regulon was favorable for S. Typhimurium virulence. Our results indicate that the deletion of phoHSTm slightly decreases the early intestinal colonization but does not affect the systemic infection of mice by S. Typhimurium (Fig. 8A and B). We observed similar phenotypes with the deletion of the siiE gene (Fig. 8), which encodes a giant nonfimbrial adhesin shown to be necessary for intestinal colonization (41, 42). In contrast, the deletion of phoHSTm did not affect the S. Typhimurium invasion of HeLa cells or RAW264.7 mouse macrophages (data not shown). Further studies into the possible role of phoH in S. Typhimurium pathogenicity are required. Even when phoH is conserved in many bacteria, its biological function remains unknown in all of them.
FIG 8.
Effect of phoHSTm in the intestinal colonization and systemic infection of mice by S. Typhimurium. Groups of 12 streptomycin-pretreated mice were orally inoculated with the WT S. Typhimurium strain or its derivative ΔphoH or ΔsiiE mutant. (A) Numbers of CFU/g from feces were determined at 1, 2, and 3 days postinfection (dpi). (B) Numbers of CFU/g from the spleen were determined at 1 day postinfection. Bars denote the standard errors of the averages for each experimental group. Statistically different values are indicated (*, P < 0.05; **, P < 0.01; ***, P < 0.001).
We found an additional trait for phoHSTm that was also generated by cis-regulatory evolution: the expression of this gene was induced by low Pi independently of the PhoR/B two-component system. A previous report indicates that only a small number of the S. Typhimurium genes that are induced by low Pi belong to the PhoB/R system regulon (43). Thus, S. Typhimurium seems to have a regulatory system alternative to PhoB/R to active phoHSTm and many other genes in response to Pi starvation. Our results indicate that HilD, as well as PhoP and SsrB, two transcriptional regulators that induce expression of Salmonella virulence genes in N-MM containing low Pi, are not involved in the activation of phoHSTm by low Pi (data not shown). Transcriptomic studies have shown that the expression of phoHSTm is induced by various types of stress that S. Typhimurium faces inside hosts, such as high concentrations of NaCl, low Fe2+, anaerobiosis, and exposure to peroxide or nitric oxide, among others (25). Various regulators could coordinate to activate the expression of phoHSTm in response to different cues, which remains to be determined.
Beyond examples for specific genes, differential regulation of multiple genes mediated by a transcriptional regulator has been described among closely related bacteria. For instance, the SlyA and Ler regulons are different between S. Typhimurium and E. coli (44) and between enteropathogenic and enterohemorrhagic E. coli strains (45). Moreover, mechanisms underlying transcriptional control evolved from prokaryotes to eukaryotes (46).
MATERIALS AND METHODS
Bacterial strains and growth conditions.
Bacterial strains used in this study are shown in Table 1. Bacterial cultures for the determination of chloramphenicol acetyltransferase (CAT) activity or for the Western blot assays were grown in 250-mL flasks containing 50 mL of LB or N-minimal medium (N-MM), which were inoculated with a 100-fold dilution of an overnight culture, as described previously (2, 47). To evaluate the low and high Pi conditions, KH2PO4 in N-MM was used at concentrations of 64 μM and 640 μM, respectively. The antibiotics ampicillin (200 μg/mL), streptomycin (100 μg/mL), and kanamycin (20 μg/mL) were added to growth media as needed.
Construction of plasmids.
Plasmids and primers used in this study are listed in Tables 1 and 2, respectively. To construct the pphoHEc-cat plasmid, the intergenic sequence upstream phoHEc was amplified by PCR with the primer pair phoH-Fw41/phoH-Rv42, using chromosomal DNA from the WT E. coli K-12 MG1655 strain as the template. To construct the pP1phoHSTm-cat, pP2phoHSTm-cat, and pphoHSTm-248 + 12-cat plasmids, the corresponding sequence upstream of phoHSTm (−653 to +12, −5 to +416, and −248 to +12, respectively, with respect to the P1 promoter) was amplified by PCR with the primer pairs phoH-Fw/phoH-Rv14, phoH-Fw15/phoH-Rv, and phoH-Fw27/phoH-Rv14, respectively, using chromosomal DNA from WT S. Typhimurium SL1344 strain as the template. The pphoHHilDmut-cat and pphoHP1mut-cat plasmids, carrying mutations in the predicted HilD-binding site and the P1 promoter, respectively, were constructed by overlapping PCR using chromosomal DNA from the WT S. Typhimurium strain as the template. First, two PCR products were obtained for each plasmid, with the primer pairs phoH-Fw/phoH-Rv80 and phoH-Fw79/phoH-Rv for pphoHHilDmut-cat and with the primer pairs phoH-Fw/phoH-Rv84 and phoH-Fw83/phoH-Rv for pphoHP1mut-cat. The PCR products were purified, and those for each plasmid were mixed and used as the template for a second PCR with the external primers phoH-Fw and phoH-Rv. To construct the ppstSSTm-cat plasmid, the intergenic sequence upstream of pstSSTm was amplified by PCR with the primer pair pstS-Fw71/pstS-Rv72, using chromosomal DNA from WT S. Typhimurium SL1344 strain as the template. All the PCR products were purified and digested whit BamHI and HindIII enzymes and then cloned into the same restriction sites of the pKK232-8 vector, which carries a promoterless cat gene (48). The pphoHSTm−67 + 3-cat plasmid was constructed with the complementary primers phoH-Fw37 and phoH-Rv38. Briefly, these primers were mixed in a 1:1 ratio, heated to 95°C for 10 min, and then cooled to room temperature for 1 h. The obtained double-strand product carries cohesive ends for its cloning into the BamHI and HindIII restriction sites of the pKK232-8 vector (48).
TABLE 2.
Primers used in this study
| Primer | Sequencea (5′–3′) | Target gene | REb |
|---|---|---|---|
| For cat transcriptional fusion | |||
| phoH-Fw41 | CGAGGATCCTGGATATGGGGTTGCTGTTTG | phoHEc | BamHI |
| phoH-Rv42 | CGAAAGCTTCATGGAGAGCACCTTGAGTTG | phoHEc | HindIII |
| phoH-Fw | CGAGGATCCAATATGGCTGGCTGGATCTG | phoHS Tm | BamHI |
| phoH-Rv14 | CGAAAGCTTGTCAGTCTCTTACAGAAAGATTAC | phoHS Tm | HindIII |
| phoH-Fw15 | CGAGGATCCTGTAAGAGACTGACAATGACGC | phoHS Tm | BamHI |
| phoH-Rv | CGAAAGCTTCCATGGATAGCACCTTGAGT | phoHS Tm | HindIII |
| phoH-Fw27 | CGAGGATCCTGACGCAATAGAGTAATGACAAAA | phoHS Tm | BamHI |
| phoH-Fw37 | GATCCCCCGCAGTAGCTAATGATTATCTTTTTTAGTCTCCTGCCGATGAAATAATCGTGTAATCTTTCTGTAAGAA | phoHS Tm | |
| phoH-Rv38 | AGCTTTCTTACAGAAAGATTACACGATTATTTCATCGGCAGGAGACTAAAAAAGATAATCATTAGCTACTGCGGGG | phoHS Tm | |
| phoH-Fw79 | CAGCATTAATACCCGCAGTAGAAAGCACACAGCTTTTTTAGTCTCCTGCCGATG | phoHS Tm | |
| phoH-Rv80 | CATCGGCAGGAGACTAAAAAAGCTGTGTGCTTTCTACTGCGGGTATTAATGCTG | phoHS Tm | |
| phoH-Fw83 | CTGCCGATGAAATAATCGTGCGGCTCTTCTGTAAGAGACTGACAATG | phoHS Tm | |
| phoH-Rv84 | CATTGTCAGTCTCTTACAGAAGAGCCGCACGATTATTTCATCGGCAG | phoHS Tm | |
| phoH-Fw17 | CGAGGATCCAGCGATGGGAGAGAGGACAC | phoHS Tm | BamHI |
| pstS-Fw71 | CGAGGATCCCTCTTTGTCCTGGCGATCCC | pstSS Tm | BamHI |
| pstS-Rv72 | CGAAAGCTTGAACAGGCCTTCCTGATTCAG | pstSS Tm | HindIII |
| For gene deletion | |||
| phoH-H1P1 | GGCTTACAAGGAAGCCAACCCTCAGATGTGTGTGCGCATAATTGTAGGCTGGAGCTGCTTCG | phoHS Tm | |
| phoH-H2P2 | CGGGCTCTGAAAGTCAATGCTATATTAGCTGTATGCGTGAAGCATATGAATATCCTCCTTAG | phoHS Tm | |
| siiE-H1P1 | ATGGGAAATAAAAGCATACAAAAGTTTTTTGCCGATCAAAATTGTAGGCTGGAGCTGCTTCG | siiE | |
| siiE-H2P2 | TTATGCGTGTTCTTCTTGATTATCTACAGGTAGCGTAACTTCCATATGAATATCCTCCTTAG | siiE | |
| SphoB H1P1 | CGACTAACAGGGCAAATTATGGCGAGACGTATTCTGGTCGTATGTAGGCTGGAGCTGCTTCG | phoBS Tm | |
| SphoB H2P2 | CAACTGGCGGAAAAGGCATTAAAAGCGGGTCGAAAAACGATACATATGAATATCCTCCTTAG | phoBS Tm | |
| For gene FLAG tagging | |||
| phoHSFlagFw | GCGCTCGGCGCTTTGTCAGCGAACGCTTCACGCATACAGCGACTACAAAGACCATGACGGT | phoHS Tm | |
| phoHSFlagRv | ACAAAGCCCGGTTTCCCGGGCTCTGAAAGTCAATGCTATACATATGAATATCCTCCTTAGTTC | phoHS Tm | |
| phoHEcFlagFw | CGTTCGGCACTTTGCCAACGTACGCTGCATGCCTACAGTGACTACAAAGACCATGACGGT | phoHEc | |
| phoHEcFlagRv | ACAAAGCCCGGTTCGCCCGGGCTCTGCACCGATAACACACCATATGAATATCCTCCTTAGTTC | phoHEc |
Underlined letters indicate the respective restriction enzyme site in the primer. The sequences corresponding to the template pKD4 or pSUB11 plasmids (Table 1) are in italics.
RE, restriction enzyme for which a site was generated in the primer.
Construction of chromosomal deletion and 3×FLAG-tagged mutant strains.
The phoB, phoH, and siiE genes were replaced with a selectable kanamycin resistance cassette in the S. Typhimurium SL1344 strain by the λRed recombinase system, as reported previously (49), using the primers shown in Table 2, generating the DTM144, DTM150, and DTM151 strains, respectively. The chromosomal phoH gene was 3×FLAG-tagged in the WT S. Typhimurium SL1344 strain as well as in the JPTM25, DTM145, MG1655, and DTM148 strains, using a previously reported method based on the λRed recombinase system (50), and the primers are shown in Table 2, generating the DTM142, DTM143, DTM146, DTM147, and DTM149 strains, respectively. The DTM145 and DTM148 strains were generated by removing the kanamycin resistance cassette from the DTM144 and JW0389 strains, respectively, using the pFLP3 plasmid expressing the FLP recombinase, as described previously (51). All modified strains were verified by PCR amplification and sequencing.
CAT assays.
The assays for CAT-specific activity were performed as described previously (52).
Western blotting.
Western blot assays were conducted as described previously (47), with minor modifications. Briefly, to detect PhoH-FLAG, sonicated cell extracts were prepared from samples of bacterial cultures grown in LB or N-MM at the indicated time. Proteins were separated on SDS–15% polyacrylamide gels. Immunoblots were performed with monoclonal anti-FLAG M2 (Sigma) or polyclonal anti-GroEL (StressGen) antibodies at 1:5,000 and 1:100,000 dilutions, respectively. To detect MBP-HilD or H-NS-FH in the EMSAs, monoclonal anti-MBP (Sigma) or monoclonal anti-FLAG M2 (Sigma) antibodies were used at a dilution of 1:3,000. Horseradish peroxidase-conjugated anti-mouse and anti-rabbit (Pierce) diluted at 1:10,000 were used as secondary antibodies. Reaction bands on membranes were developed with the Western Lightning chemiluminescence reagent plus (Perkin-Elmer) and exposition to KodaK X-Omat films.
Sequence analysis.
BLASTP and Clustal Omega2 (EMBL-EBI) were used to compare amino acid and nucleotide sequences, respectively. The phylogenetic tree based on the P1phoH−67 + 3 sequence was generated with the IQ-TREE (53) and interactive Tree of Life (v6) (54) software using default parameters. Prediction of HilD-binding sites was performed with the previously reported position-specific scoring matrices, representing HilD-binding consensus sequences (10), using the matrix-scan program (55) with a P value of 1e−3. Prediction of PhoB-binding site was accomplished with the BPROM tool of the Softberry software (56).
Expression and purification of MBP-HilD and H-NS-FH.
The maltose binding protein (MBP)-HilD and the H-NS-FLAG-His6 (H-NS-FH) protein were expressed in the E. coli BL21/DE3 strain containing the pMAL-HilD1 or pBAD-H-NS-FH plasmid and purified by using amylose-agarose or Ni2+-nitrilotriacetic-agarose affinity columns, respectively, as described previously (2).
EMSAs.
The fragments P1 and P2, spanning the phoHSTm sequences contained in the P1phoHSTm-cat and P2phoHSTm-cat fusions, were obtained by PCR amplification using the primer pairs phoH-Fw/phoH-Rv14 and phoH-Fw15/phoH-Rv, respectively, and chromosomal DNA from the WT S. Typhimurium SL1344 strain as the template. The WT and mutated P1phoH-157/+12 fragments were obtained by PCR using the primer pairs phoH-Fw17/phoH-Rv14 and phoH-Fw17/phoH-Rv84 and the DNA from the pphoHSTm-cat (WT), pphoHHilDmut-cat (mutations in the predicted HilD-binding site), or pphoHP1mut-cat (mutations in the P1 promoter) plasmids as the template. The fragments spanning the phoHSTm or phoHEc full-length regulatory region, contained in the phoHSTm-cat or phoHEc-cat fusions, were obtained by PCR using the primer pairs phoH-Fw/phoH-Rv and phoH-Fw41/phoH-Rv42 and chromosomal DNA from WT S. Typhimurium SL1344 and E. coli MG1655 strains as the template, respectively. The final PCR products were purified with the Zymo Research DNA Clean & Concentrator kit (ZYMO RESARCH). Protein-DNA binding reactions with purified MBP-HilD and/or H-NS-FH proteins were analyzed as described previously (2, 57). For competitive EMSAs, the WT P1phoH-157/+12 fragment was first incubated with 4 μM H-NS-FH for 15 min and then incubated with increasing concentrations of MBP-HilD for an additional 20 min. Binding mixtures were electrophoretically separated in 6% nondenaturing acrylamide in 0.5× Tris-borate-EDTA buffer at room temperature. DNA bands were visualized by staining with ethidium bromide in an Alpha-Imager UV transilluminator (Alpha Innotech Corp.). MBP-HilD and H-NS-FLAG were detected on the DNA/protein complexes by Western blotting.
Mouse infection assays.
Mouse experiments were conducted according to the standard operating protocols approved by the International Committee for Animal Care and Use from CICUAL-UNAM and by the Official Mexican Norm NOM-062-Z00-1999. Pathogen-free BALB/c female mice (6 to 7 weeks old) were obtained from the Experimental Medicine Research Unit, School of Medicine, UNAM, Mexico. Maintenance, treatment with 50 mg of streptomycin, infection, and euthanasia of the mice, as well as the counting of the number of CFU per gram from feces or the spleen were performed as described previously (57, 58). Groups of 12 streptomycin-pretreated mice were infected by the orogastric route with bacterial suspensions containing 1.0 × 106 CFU of the WT S. Typhimurium strain or its derivative ΔphoH and ΔsiiE mutants, prepared in sterile 1× phosphate-buffered saline.
Cell infection assays.
Invasion of HeLa cells or RAW264.7 mouse macrophages was tested by gentamicin assays, as described previously (57, 59).
Statistical analysis.
Statistical analyses were performed with the GraphPad Prism 6.0 or 9.2.0 software (GraphPad Inc., San Diego, CA), using the one-way analysis of variance (ANOVA) or Student's t test (unpaired). P values of <0.05 were considered statistically significant.
ACKNOWLEDGMENTS
This work was supported by grants from Dirección General de Asuntos del Personal Académico de la UNAM/México (IN202418 and IN206321) and Consejo Nacional de Ciencia y Tecnología (CONACYT)/México (254531) to V.H.B. and from CONACYT/México (256263) to M.A.D.C. M.A.V.D. was supported by a predoctoral fellowship from CONACYT (594831).
The funders had no role in the study design, data collection and interpretation, or the decision to submit the work for publication.
We thank M. Fernández-Mora and F. J. Santana for technical assistance, H. Salgado and I. Martínez-Flores for prediction of HilD-binding sites, M. M. Banda for constructing the ΔsiiE mutant, and I. Martínez-Flores and D. Pérez-Morales for critical reading of the manuscript.
Contributor Information
Víctor H. Bustamante, Email: victor.bustamante@ibt.unam.mx.
George O'Toole, Geisel School of Medicine at Dartmouth.
REFERENCES
- 1.Wang L, Wang FF, Qian W. 2011. Evolutionary rewiring and reprogramming of bacterial transcription regulation. J Genet Genomics 38:279–288. 10.1016/j.jgg.2011.06.001. [DOI] [PubMed] [Google Scholar]
- 2.Bustamante VH, Martínez LC, Santana FJ, Knodler LA, Steele-Mortimer O, Puente JL. 2008. HilD-mediated transcriptional cross-talk between SPI-1 and SPI-2. Proc Natl Acad Sci USA 105:14591–14596. 10.1073/pnas.0801205105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fàbrega A, Vila J. 2013. Salmonella enterica serovar Typhimurium skills to succeed in the host: virulence and regulation. Clin Microbiol Rev 26:308–341. 10.1128/CMR.00066-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Perez JC, Groisman EA. 2009. Evolution of transcriptional regulatory circuits in bacteria. Cell 138:233–244. 10.1016/j.cell.2009.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ilyas B, Tsai CN, Coombes BK. 2017. Evolution of Salmonella-host cell interactions through a dynamic bacterial genome. Front Cell Infect Microbiol 7:428. 10.3389/fcimb.2017.00428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fookes M, Schroeder GN, Langridge GC, Blondel CJ, Mammina C, Connor TR, Seth-Smith H, Vernikos GS, Robinson KS, Sanders M, Petty NK, Kingsley RA, Bäumler AJ, Nuccio S-P, Contreras I, Santiviago CA, Maskell D, Barrow P, Humphrey T, Nastasi A, Roberts M, Frankel G, Parkhill J, Dougan G, Thomson NR. 2011. Salmonella bongori provides insights into the evolution of the Salmonellae. PLoS Pathog 7:e1002191. 10.1371/journal.ppat.1002191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lostroh CP, Lee CA. 2001. The Salmonella pathogenicity island-1 type III secretion system. Microbes Infect 3:1281–1291. 10.1016/s1286-4579(01)01488-5. [DOI] [PubMed] [Google Scholar]
- 8.Que F, Wu S, Huang R. 2013. Salmonella pathogenicity island 1(SPI-1) at work. Curr Microbiol 66:582–587. 10.1007/s00284-013-0307-8. [DOI] [PubMed] [Google Scholar]
- 9.Schechter LM, Lee CA. 2001. AraC/XylS family members, HilC and HilD, directly bind and derepress the Salmonella typhimurium hilA promoter. Mol Microbiol 40:1289–1299. 10.1046/j.1365-2958.2001.02462.x. [DOI] [PubMed] [Google Scholar]
- 10.Martínez-Flores I, Pérez-Morales D, Sánchez-Pérez M, Paredes CC, Collado-Vides J, Salgado H, Bustamante VH. 2016. In silico clustering of Salmonella global gene expression data reveals novel genes co-regulated with the SPI-1 virulence genes through HilD. Sci Rep 6:37858. 10.1038/srep37858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Smith C, Stringer AM, Mao C, Palumbo MJ, Wade JT. 2016. Mapping the regulatory network for Salmonella enterica serovar Typhimurium invasion. mBio 7:e01024-16. 10.1128/mBio.01024-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Colgan AM, Kröger C, Diard M, Hardt WD, Puente JL, Sivasankaran SK, Hokamp K, Hinton JCD. 2016. The impact of 18 ancestral and horizontally-acquired regulatory proteins upon the transcriptome and sRNA landscape of Salmonella enterica serovar Typhimurium. PLoS Genet 12:e1006258-42. 10.1371/journal.pgen.1006258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Petrone BL, Stringer AM, Wade JT. 2014. Identification of HilD-regulated genes in Salmonella enterica serovar Typhimurium. J Bacteriol 196:1094–1101. 10.1128/JB.01449-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Singer HM, Kühne C, Deditius JA, Hughes KT, Erhardt M. 2014. The Salmonella Spi1 virulence regulatory protein HilD directly activates transcription of the flagellar master operon flhDC. J Bacteriol 196:1448–1457. 10.1128/JB.01438-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mouslim C, Hughes KT. 2014. The effect of cell growth phase on the regulatory cross-talk between flagellar and Spi1 virulence gene expression. PLoS Pathog 10:e1003987. 10.1371/journal.ppat.1003987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Banda MM, Manzo R, Bustamante VH. 2019. HilD induces expression of a novel Salmonella Typhimurium invasion factor, YobH, through a regulatory cascade involving SprB. Sci Rep 9:12725. 10.1038/s41598-019-49192-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rivera-Chávez F, Winter SE, Lopez CA, Xavier MN, Winter MG, Nuccio SP, Russell JM, Laughlin RC, Lawhon SD, Sterzenbach T, Bevins CL, Tsolis RM, Harshey R, Adams LG, Bäumler AJ. 2013. Salmonella uses energy taxis to benefit from intestinal inflammation. PLoS Pathog 9:e1003267. 10.1371/journal.ppat.1003267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Schmitt CK, Ikeda JS, Darnell SC, Watson PR, Bispham J, Wallis TS, Weinstein DL, Metcalf ES, O'Brien AD. 2001. Absence of all components of the flagellar export and synthesis machinery differentially alters virulence of Salmonella enterica serovar Typhimurium in models of typhoid fever, survival in macrophages, tissue culture invasiveness, and calf enterocolitis. Infect Immun 69:5619–5625. 10.1128/IAI.69.9.5619-5625.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stecher B, Hapfelmeier S, Müller C, Kremer M, Stallmach T, Hardt W-D. 2004. Flagella and chemotaxis are required for efficient induction of Salmonella enterica serovar Typhimurium colitis in streptomycin-pretreated mice. Infect Immun 72:4138–4150. 10.1128/IAI.72.7.4138-4150.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Horstmann JA, Lunelli M, Cazzola H, Heidemann J, Kühne C, Steffen P, Szefs S, Rossi C, Lokareddy RK, Wang C, Lemaire L, Hughes KT, Uetrecht C, Schlüter H, Grassl GA, Stradal TEB, Rossez Y, Kolbe M, Erhardt M. 2020. Methylation of Salmonella Typhimurium flagella promotes bacterial adhesion and host cell invasion. Nat Commun 11:2013. 10.1038/s41467-020-15738-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhou L, Lei XH, Bochner BR, Wanner BL. 2003. Phenotype MicroArray analysis of Escherichia coli K-12 mutants with deletions of all two-component systems. J Bacteriol 185:4956–4972. 10.1128/JB.185.16.4956-4972.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kazakov AE, Vassieva O, Gelfand MS, Osterman A, Overbeek R. 2003. Bioinformatics classification and functional analysis of PhoH homologs. In Silico Biol 3:3–15. [PubMed] [Google Scholar]
- 23.Kim SK, Makino K, Amemura M, Shinagawa H, Nakata A. 1993. Molecular analysis of the phoH gene, belonging to the phosphate regulon in Escherichia coli. J Bacteriol 175:1316–1324. 10.1128/jb.175.5.1316-1324.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Santos-Beneit F. 2015. The Pho regulon: a huge regulatory network in bacteria. Front Microbiol 6:402. 10.3389/fmicb.2015.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kröger C, Colgan A, Srikumar S, Händler K, Sivasankaran SK, Hammarlöf DL, Canals R, Grissom JE, Conway T, Hokamp K, Hinton JCD. 2013. An infection-relevant transcriptomic compendium for Salmonella enterica serovar Typhimurium. Cell Host Microbe 14:683–695. 10.1016/j.chom.2013.11.010. [DOI] [PubMed] [Google Scholar]
- 26.Ramachandran VK, Shearer N, Thompson A. 2014. The primary transcriptome of Salmonella enterica serovar Typhimurium and its dependence on ppGpp during late stationary phase. PLoS One 9:e92690. 10.1371/journal.pone.0092690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Olekhnovich IN, Kadner RJ. 2007. Role of nucleoid-associated proteins Hha and H-NS in expression of Salmonella enterica activators HilD, HilC, and RtsA required for cell invasion. J Bacteriol 189:6882–6890. 10.1128/JB.00905-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Martínez LC, Banda MM, Fernández-Mora M, Santana FJ, Bustamante VH. 2014. HilD induces expression of Salmonella pathogenicity island 2 genes by displacing the global negative regulator H-NS from ssrAB. J Bacteriol 196:3746–3755. 10.1128/JB.01799-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Banda MM, Zavala-Alvarado C, Pérez-Morales D, Bustamante VH. 2019. SlyA and HilD counteract H-NS-mediated repression on the ssrAB virulence operon of Salmonella enterica serovar Typhimurium and thus promote its activation by OmpR. J Bacteriol 201:e00530-18. 10.1128/JB.00530-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Romero-González LE, Pérez-Morales D, Cortés-Avalos D, Vázquez-Guerrero E, Paredes-Hernández DA, Estrada-De los Santos P, Villa-Tanaca L, de la Cruz MA, Bustamante VH, Antonio Ibarra J. 2020. The Salmonella Typhimurium InvF-SicA complex is necessary for the transcription of sopB in the absence of the repressor H-NS. PLoS One 15:e0240617-18. 10.1371/journal.pone.0240617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ali SS, Soo J, Rao C, Leung AS, Ngai DHM, Ensminger AW, Navarre WW. 2014. Silencing by H-NS potentiated the evolution of Salmonella. PLoS Pathog 10:e1004500. 10.1371/journal.ppat.1004500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Navarre WW, Porwollik S, Wang Y, McClelland M, Rosen H, Libby SJ, Fang FC. 2006. Selective silencing of foreign DNA with low GC content by the H-NS protein in Salmonella. Science 313:236–238. 10.1126/science.1128794. [DOI] [PubMed] [Google Scholar]
- 33.Fang FC, Rimsky S. 2008. New insights into transcriptional regulation by H-NS. Curr Opin Microbiol 11:113–120. 10.1016/j.mib.2008.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yang C, Huang TW, Wen SY, Chang CY, Tsai SF, Wu WF, Chang CH. 2012. Genome-wide PhoB binding and gene expression profiles reveal the hierarchical gene regulatory network of phosphate starvation in Escherichia coli. PLoS One 7:e47314. 10.1371/journal.pone.0047314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Winfield MD, Groisman EA. 2004. Phenotypic differences between Salmonella and Escherichia coli resulting from the disparate regulation of homologous genes. Proc Natl Acad Sci USA 101:17162–17167. 10.1073/pnas.0406038101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Osborne SE, Walthers D, Tomljenovic AM, Mulder DT, Silphaduang U, Duong N, Lowden MJ, Wickham ME, Waller RF, Kenney LJ, Coombes BK. 2009. Pathogenic adaptation of intracellular bacteria by rewiring a cis-regulatory input function. Proc Natl Acad Sci USA 106:3982–3987. 10.1073/pnas.0811669106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ilyas B, Mulder DT, Little DJ, Elhenawy W, Banda MM, Pérez-Morales D, Tsai CN, Chau NYE, Bustamante VH, Coombes BK. 2018. Regulatory evolution drives evasion of host inflammasomes by Salmonella Typhimurium. Cell Rep 25:825–832. 10.1016/j.celrep.2018.09.078. [DOI] [PubMed] [Google Scholar]
- 38.Choi J, Groisman EA. 2020. Horizontally acquired regulatory gene activates ancestral regulatory system to promote Salmonella virulence. Nucleic Acids Res 48:10832–10847. 10.1093/nar/gkaa813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Olekhnovich IN, Kadner RJ. 2006. Crucial roles of both flanking sequences in silencing of the hilA promoter in Salmonella enterica. J Mol Biol 357:373–386. 10.1016/j.jmb.2006.01.007. [DOI] [PubMed] [Google Scholar]
- 40.Boddicker JD, Knosp BM, Jones BD. 2003. Transcription of the Salmonella invasion gene activator, hilA, requires HilD activation in the absence of negative regulators. J Bacteriol 185:525–533. 10.1128/JB.185.2.525-533.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Morgan E, Campbell JD, Rowe SC, Bispham J, Stevens MP, Bowen AJ, Barrow PA, Maskell DJ, Wallis TS. 2004. Identification of host-specific colonization factors of Salmonella enterica serovar Typhimurium. Mol Microbiol 54:994–1010. 10.1111/j.1365-2958.2004.04323.x. [DOI] [PubMed] [Google Scholar]
- 42.Gerlach RG, Jäckel D, Stecher B, Wagner C, Lupas A, Hardt WD, Hensel M. 2007. Salmonella pathogenicity island 4 encodes a giant non-fimbrial adhesin and the cognate type 1 secretion system. Cell Microbiol 9:1834–1850. 10.1111/j.1462-5822.2007.00919.x. [DOI] [PubMed] [Google Scholar]
- 43.Jiang J, Yu K, Qi L, Liu Y, Cheng S, Wu M, Wang Z, Fu J, Liu X. 2018. A proteomic view of Salmonella Typhimurium in response to phosphate limitation. Proteomes 6:19. 10.3390/proteomes6020019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Spory A, Bosserhoff A, Von Rhein C, Goebel W, Ludwig A. 2002. Differential regulation of multiple proteins of Escherichia coli and Salmonella enterica serovar typhimurium by the transcriptional regulator SlyA. J Bacteriol 184:3549–3559. 10.1128/JB.184.13.3549-3559.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bingle LEH, Constantinidou C, Shaw RK, Islam MS, Patel M, Snyder LAS, Lee DJ, Penn CW, Busby SJW, Pallen MJ. 2014. Microarray analysis of the Ler regulon in enteropathogenic and enterohaemorrhagic Escherichia coli strains. PLoS One 9:e80160. 10.1371/journal.pone.0080160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bylino OV, Ibragimov AN, Shidlovskii YV. 2020. Evolution of regulated transcription. Cells 9:1675–1678. 10.3390/cells9071675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Martínez LC, Yakhnin H, Camacho MI, Georgellis D, Babitzke P, Puente JL, Bustamante VH. 2011. Integration of a complex regulatory cascade involving the SirA/BarA and Csr global regulatory systems that controls expression of the Salmonella SPI-1 and SPI-2 virulence regulons through HilD. Mol Microbiol 80:1637–1656. 10.1111/j.1365-2958.2011.07674.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Brosius J. 1984. Plasmid vectors for the selection of promoters. Gene 27:151–160. 10.1016/0378-1119(84)90136-7. [DOI] [PubMed] [Google Scholar]
- 49.Datsenko KA, Wanner BL. 2000. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci USA 97:6640–6645. 10.1073/pnas.120163297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Uzzau S, Figueroa-Bossi N, Rubino S, Bossi L. 2001. Epitope tagging of chromosomal genes in Salmonella. Proc Natl Acad Sci USA 98:15264–15269. 10.1073/pnas.261348198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Choi KH, Gaynor JB, White KG, Lopez C, Bosio CM, Karkhoff-Schweizer RAR, Schweizer HP. 2005. A Tn7-based broad-range bacterial cloning and expression system. Nat Methods 2:443–448. 10.1038/nmeth765. [DOI] [PubMed] [Google Scholar]
- 52.Puente JL, Bieber D, Ramer SW, Murray W, Schoolnik GK. 1996. The bundle-forming pili of enteropathogenic Escherichia coli: transcriptional regulation by environmental signals. Mol Microbiol 20:87–100. 10.1111/j.1365-2958.1996.tb02491.x. [DOI] [PubMed] [Google Scholar]
- 53.Nguyen LT, Schmidt HA, Von Haeseler A, Minh BQ. 2015. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol 32:268–274. 10.1093/molbev/msu300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Letunic I, Bork P. 2021. Interactive Tree Of Life (iTOL) v5: an online tool for phylogenetic tree display and annotation. Nucleic Acids Res 49:W293–W296. 10.1093/nar/gkab301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Medina-Rivera A, Defrance M, Sand O, Herrmann C, Castro-Mondragon JA, Delerce J, Jaeger S, Blanchet C, Vincens P, Caron C, Staines DM, Contreras-Moreira B, Artufel M, Charbonnier-Khamvongsa L, Hernandez C, Thieffry D, Thomas-Chollier M, Van Helden J. 2015. RSAT 2015: regulatory sequence analysis tools. Nucleic Acids Res 43:W50–W56. 10.1093/nar/gkv362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Solovyev V, Salamov A. 2011. Automatic annotation of microbial genomes and metagenomic sequences, p 69–78. In Li RW (ed), Metagenomics and its applications in agriculture, biomedicine and environmental studies. Nova Science Publishers, Hauppauge, NY. [Google Scholar]
- 57.Banda MM, López C, Manzo R, Rico-Pérez G, García P, Rosales-Reyes R, De la Cruz MA, Soncini FC, García-del Portillo F, Bustamante VH. 2018. HilD and PhoP independently regulate the expression of grhD1, a novel gene required for Salmonella Typhimurium invasion of host cells. Sci Rep 8:4841. 10.1038/s41598-018-23068-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Pérez-Morales D, Nava-Galeana J, Rosales-Reyes R, Teehan P, Yakhnin H, Melchy-Pérez EI, Rosenstein Y, De la Cruz MA, Babitzke P, Bustamante VH. 2021. An incoherent feedforward loop formed by SirA/BarA, HilE and HilD is involved in controlling the growth cost of virulence factor expression by Salmonella Typhimurium. PLoS Pathog 17:e1009630. 10.1371/journal.ppat.1009630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pérez-Morales D, Banda MM, Chau NYE, Salgado H, Martínez-Flores I, Ibarra JA, Ilyas B, Coombes BK, Bustamante VH. 2017. The transcriptional regulator SsrB is involved in a molecular switch controlling virulence lifestyles of Salmonella. PLoS Pathog 13:e1006497-27. 10.1371/journal.ppat.1006497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hoiseth SK, Stocker BA. 1981. Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines. Nature 291:238–239. 10.1038/291238a0. [DOI] [PubMed] [Google Scholar]
- 61.Ferenci T, Zhou Z, Betteridge T, Ren Y, Liu Y, Feng L, Reeves PR, Wang L. 2009. Genomic sequencing reveals regulatory mutations and recombinational events in the widely used MC4100 lineage of Escherichia coli K-12. J Bacteriol 191:4025–4029. 10.1128/JB.00118-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bustamante VH, Villalba MI, García-Angulo VA, Vázquez A, Martínez LC, Jiménez R, Puente JL. 2011. PerC and GrlA independently regulate Ler expression in enteropathogenic Escherichia coli. Mol Microbiol 82:398–415. 10.1111/j.1365-2958.2011.07819.x. [DOI] [PubMed] [Google Scholar]
- 63.Baba T, Ara T, Hasegawa M, Takai Y, Okumura Y, Baba M, Datsenko KA, Tomita M, Wanner BL, Mori H. 2006. Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol Syst Biol 10.1038/msb4100050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mayer MP. 1995. A new set of useful cloning and expression vectors derived from pBlueScript. Gene 163:41–46. 10.1016/0378-1119(95)00389-n. [DOI] [PubMed] [Google Scholar]








