Introduction

Edwardsiella was isolated from infected humans and animals and identified as a new genus of Enterobacteriaceae in 1965 [1]. Recently, the Edwardsiella genus was classified into five species, including E. tarda, E. anguillarum, E. ictaluri, E. hoshinae, and E. piscicida [2, 3]. E. tarda (now is also considered to be E. piscicida), a member of the family Enterobacteriaceae, is a gram-negative, motile, facultative anaerobe and rod-shaped bacterium [4, 5]. E. tarda has broad host ranges including aquatic animals, reptiles, amphibians, birds, mammals and humans [6,7,8], therefore being a significant zoonotic pathogen [9]. This pathogen can cause serious systemic infections and high mortality in both seawater and freshwater fish, accounting for severe economic losses and heavily influencing the healthy development of aquaculture [10, 14, 15]. These acidic environments include acid soil and fermented food, the harsh inorganic and organic volatile fatty environment in the gastrointestinal tract, as well as mild to moderate acids in phagosomes of cell macrophages [16]. Therefore, an important strategy for pathogens is to adapt to the acidic stress environment of the host. To counter intracellular and extracellular acid stresses, bacteria have developed passive and active protective mechanisms [17]. Among these mechanisms, the amino acid-dependent acid resistance (AR) system is composed of a cytoplasmic decarboxylase that catalyzes a proton-dependent decarboxylation of a substrate amino acid, and an inner membrane substrate/product antiporter that exchanges external substrate for internal product [18]. In E. coli, the AR systems that have been identified are the glutamic acid-dependent acid resistance (GDAR) system [19], the arginine-dependent acid resistance (ADAR) system [20], the lysine-dependent acid resistance (LDAR) system [21] and the ornithine-dependent acid resistance (ODAR) system [22]. Each of these systems has a different optimum pH and is capable of providing protection against acid stress over a broad range of pH values [23].

The LDAR system is a mild acid tolerance response system. In E. coli, the LDAR system consists of the cytoplasmic protein CadA, the integral membrane proteins CadB, and one-component regulator CadC [24]. CadA, the inducible lysine decarboxylase, is encoded by cadA gene that converts lysine to cadaverine with consumption of a proton [25, 26]. CadB, the lysine/cadaverine antiporter with 12 transmembrane helices, is encoded by the cadB gene that transports lysine into the cell and exports cadaverine. cadB and cadA are organized in one operon which work together to maintain pH homeostasis inside the cell and extracellular environment [27]. CadC is a membrane-integrated transcription activator located upstream of cadB. The regulator belongs to the ToxR family, consisting of a cytoplasmic N-terminal DNA-binding domain that regulates transcription, a transmembrane helix, and a C-terminal periplasmic sensory domain [28]. Under conditions of acidic pH and exogenous lysine, CadC interaction with the lysine-specific permease LysP senses signal transduction to the DNA-binding domain via the transmembrane helix [29]. DNA-binding domain undergoes structural changes that enable the winged protein to interact with the target promoter of cadBA and activated expression of the cadBA operon [30]. The LDAR system has been thoroughly studied in Enterobacteria such as E. coli [31], Salmonella typhimurium [15] and Vibrio [32] because of direct links between the efficiency of the acid stress response and pathogenicity, but little is known about biological roles in E. tarda.

In this study, we investigated the properties of cadBA operon in E. tarda, and analyzed its distinct roles in the physiological fitness and pathogenesis in in vitro and in vivo models of infection with single and double deletion mutations in cadB and cadA. Our results uncover a vital role of cadBA operon in E. tarda survival under acidic stress environment and in host cells, and provide the first insights into the pathogenicity of the AR system in E. tarda.

Materials and methods

Strains and culture conditions

E. coli DH5ɑ was purchased from TransGen (Bei**g, China) and used for genetic manipulation. E. coli SM10 λpir or D1000 was purchased from Biomedal (Sevilla, Spain) and used for preparation of suicide vectors. E. tarda TX01 was isolated from diseased fish [33]. Bacteria were grown in Luria–Bertani (LB) broth or on LB agar at 37 °C (for E. coli) or 28 °C (for E. tarda). When required, antibiotics were added to the media as follows: ampicillin (Amp), 100 μg/mL; chloramphenicol (Cm), 30 μg/mL; polymyxin B (poly B) or colistin (Col), 100 μg/mL; tetracycline (Tc), 20 μg/mL and kanamycin (Km), 50 μg/mL, respectively.

Total RNA isolation, cDNA synthesis, and co-transcriptional verification

The experiment was performed as previously reported [34]. Overnight cultures of E. tarda TX01 were grown in LB broth at 28 °C and used for RT-PCR. Total RNA was isolated using the HP Total RNA kit (Omega Bio-Tek, USA) according to the manufacturer’s instructions. cDNA synthesis was performed with Superscript II reverse transcriptase (Invitrogen, USA). The cadBA co-transcriptional fragment was located at the 3ʹ end of cadB and 5ʹ end of cadA, and amplified with specific primers CadBAF and CadBAR using total RNA, cDNA, and genomic DNA as the template, respectively. The sequences of primers used are shown in Table 1.

Table 1 Oligonucleotide primers used in this study

The expression of acid resistance genes under acidic conditions and in cadC mutant

To study expression of acid resistance genes under exposure to acid stress conditions, the exponential phase cultures of TX01 were treated for 1 h in LB media supplemented with 5 mM l-lysine (pH 5.5) in static cultivation. Total RNA was extracted with an HP Total RNA kit (Omega Bio-Tek, USA) according to the manufacturer’s instructions. cDNA synthesis was performed as described above. RT-qPCR was carried out as reported previously [35]. The relative transcriptional level of acidic resistance genes (cadC, cadB, cadA, and cadBA) in E. tarda were determined using the 2−△△CT method with 16S rRNA used as a reference gene.

Constructions of mutant strains TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA and plasmid-complemented strains TX01ΔcadAC and TX01ΔcadBC

The constructions of mutants TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA and complementary strains TX01ΔcadAC and TX01ΔcadBC were performed as reported previously [35]. The primers used in this study are listed in Table 1. Primers CadAKOF1/CadAKOR1 and CadAKOF2/CadAKOR2 were used for the construction of TX01ΔcadA mutant, in which a 1023 bp segment (517 to 1539 bp in the ORF) in-frame deletion was created. Primers CadBKOF1/CadBKOR1 and CadBKOF2/CadBKOR2 were used for the construction of TX01ΔcadB mutant, in which a 636 bp segment (337 to 972 bp in the ORF) in-frame deletion was created. The transconjugants were selected on LB agar plates supplemented with 10% sucrose. cadA single-knockout strain was named as TX01ΔcadA, and cadB single-knockout strain was named as TX01ΔcadB. A similar operation was carried out to construct the double-knockout strain TX01ΔcadBA.

For construction of TX01ΔcadA complementary strain, a 2145-bp cadA coding region and 418-bp putative promoter region were cloned with primers CadAF1/CadAR1 and CadBAPF1/CadBAPR2, respectively. Two DNA fragments were fused together by overlap** PCR with CadBAPF1/CadAR1 and verified by sequencing. The fused PCR fragment was digested with EcoRV, ligated into plasmid pJR21 digested with PmeI. The resulting plasmid was transformed into TX01ΔcadA by conjugation transfer. The complementary strain TX01ΔcadAC was screened on LB agar plates supplemented with 100 μg/mL polymyxin B and 20 μg/mL tetracycline, and further confirmed by PCR. For construction of TX01ΔcadB complementary strain, a 1332-bp cadB coding region and a 418-bp upstream of transcriptional start site was amplified from TX01 with primers CadBAPF1/CadBR1. The following experimental operations are similar to the construction of TX01ΔcadAC. The complementary strains were named as TX01ΔcadAC and TX01ΔcadBC, respectively.

Resistance to environmental stresses

The overnight cultures of TX01, TX01ΔcadA, TX01ΔcadB and TX01ΔcadBA were grown in LB media, diluted to a final concentration of about 105 CFU/mL. For acid stress, aliquots of (200 μL) cultures in LB broth at pH gradients (from 7.0 to 4.5) were distributed into the wells of Bioscreen C plate. For the iron deficiency assay, bacteria were added into fresh LB media with 50 μM of 2, 2ʹ-dipyridyl (Dp). For oxidation stress assay, bacteria were added into fresh LB media with 500 μM of diamide. Growth curves were monitored at 2-h intervals through the measurement of the absorbance at 600 nm using Bioscreen C Automated Growth Curve System (OyGrowth Curves Ab Ltd, Finland).

Lysine decarboxylase assay

Lysine decarboxylase broth was purchased from Hopebio (Qingdao, China) and used for the lysine decarboxylase (LDC) qualitative measurement. Various overnight cultures of bacteria (TX01, TX01∆cadA, TX01∆cadB, TX01∆cadBA, TX01∆cadAC and TX01∆cadBC) were diluted to an optical density of 0.5 at 600 nm. A 50-μL aliquot of the cultures were inoculated into lysine decarboxylase broth after being washed two times and resuspended with sterile water. Sterile paraffin-coated liquid (300 μL) was then added into inoculated media to create an anaerobic environment and the cultures were incubated at 28 °C for 10, 20 and 30 h. Lysine decarboxylase broth without l-lysine was taken as a blank control. A positive test is indicated if the control tube is yellow and the LDC-positive tube is purple or red brown color. On the contrary, a negative test is indicated if both the control tube and the LDC-positive tube are yellow.

Strains were grown in LB broth at 28 °C until OD600 of 0.5. Culture aliquots were collected and normalized to an OD600 of 1.0. Quantification of lysine decarboxylase activity was then carried out as described previously [36]. Lysine decarboxylase activity is a measure of lysine converted to cadaverine per time (min) per unit cell density, and was determined by A340 between the sample incubated with or without lysine using the following equation:

$$[A340/\left( {{\text{time}}\, \times \,{\text{OD}}_{600} } \right)]\, \times \,1000.$$

Biofilm assay

Biofilm formation was assayed using crystal violet staining as previously described [37]. Overnight cultures of TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC and TX01ΔcadBC were grown in LB media, diluted to 105 CFU/mL, and then resuspended in LB media at pH 7.0 and 5.5. 200 μL of cultures were transferred into 96-well polystyrene plates and incubated at 28 °C for 24 h under static conditions. The media and planktonic cells were discarded after culture and the wells were washed three times with PBS. The adhered cells were treated with Bouin fixative (Solarbio, China) for 1 h and then stained with 1% crystal violet (CV, Sigma) solution for 20 min, followed by the removal of unbound crystal violet by washing several times with PBS. The CV bound to biofilm was solubilized with 200 μL methanol and absorbance was measured at 570 nm.

For confocal laser scanning microscopy (CLSM) observation, TX01, TX01ΔcadA, TX01ΔcadB and TX01ΔcadBA were cultured in LB broth on glass-bottom dishes at 28 °C for 24 h. Non-adherent cells and culture fluid were removed and washed three times with PBS. The biofilms were stained with a LIVE/DEAD BacLight bacterial viability kit L-13152 (Invitrogen-Molecular Probes, USA) for 15 min at room temperature in dark conditions. The fluorescent images were acquired using a FV1000 confocal laser scanning microscope (Olympus, Japan).

Motility assays

Swimming motility assays were performed using swimming plates which contain LB media with 0.3% (W/V) agar at pH 7.0 or pH 5.5. Overnight cultures of TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC and TX01ΔcadBC were cultured in LB media to an OD600 of 0.5. Aliquots of (1 μL) cell suspensions were inoculated into swimming plates by submerging pipette tips in the cultures and pricking the center of the plates. Plates were then incubated for 18 h, and the swimming motility zone was measured.

Invasion of eukaryotic cell lines

The human cervical epithelial cell line HeLa and the murine monocyte-macrophage cell line RAW264.7 were cultured in Dulbecco Modified Eagle medium (DMEM) supplemented with 10% (V/V) fetal bovine serum (FBS, Sigma), 100 U/mL penicillin G (Sigma), and 100 μg/mL streptomycin (Sigma) at 37 °C with 5% CO2.

The interaction of E. tarda and host cells was studied as reported previously [38]. Briefly, E. tarda TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC strains were grown in LB broth at 30 °C and then transferred into DMEM without shaking until the optical density at 600 nm reached 1.0. HeLa cells were seeded and cultured in 96-well cell culture plates, and infected with 100 μL of E. tarda at a multiplicity of infection (MOI) of 10:1. To detect bacterial adhesion, after incubation at 28 °C for 1 and 2 h, the monolayers were washed three times with PBS and lysed with 200 μL of 1% (V/V) Triton X-100 for 10 min; series diluted bacteria were quantified by LB agar plates supplemented with polymyxin B.

Bacterial replication in RAW264.7 cells was performed as previously [39]. Briefly, RAW264.7 cells were infected with 100 μL E. tarda strains at MOI of 10:1. The monolayers were washed with PBS and then in cultured DMEM containing 200 μg/mL of gentamicin for 2 h to kill the extracellular bacteria, then they were maintained in DMEM including 10 μg/mL of gentamicin for 1 h. Subsequently, the cultures were incubated at 28 °C for 0, 2, 4, and 8 h. The monolayers were washed three times with PBS and lysed with 1% (V/V) Triton X-100 for enumeration.

For microscopy observation, E. tarda was transformed with plasmid pGFPUV expressing green fluorescence protein by electroporation using Gemini Twin Wave Electroporators [40]. RAW264.7 cells were cultured in glass-bottom dishes and infected with TX01, TX01ΔcadA, TX01ΔcadB and TX01ΔcadBA strains possessing the pGFPUV plasmid at 30 °C for 0, 2, and 4 h. The cells were washed three times with PBS and fixed by polyformaldehyde for 30 min, and then labeled with DAPI (Solarbio, China). The cells were observed by a confocal microscope (Olympus Fluoview FV1000, Japan) after washing three times with PBS.

Fish and experimental challenges for bacterial dissemination in vivo

Healthy Tilapias (average weight 13.0 g) were purchased from a commercial fish farm in Haikou, and fed in aerated water at 25 ± 1 °C for 2 weeks. Before the experiment, the fish were randomly sampled and checked for the presence of bacteria in the blood, liver, kidney and spleen. For tissue dissemination analysis, TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC were cultured in LB broth to an OD600 of 0.5. The cells were washed with PBS and resuspended in PBS to 107 CFU/mL. Tilapia were divided randomly into six groups and infected by intramuscular (i. m.) injection with 50 μL of various bacterial suspensions, respectively. Fish were euthanized with an overdose of MS222 (tricaine methanesulfonate) (Sigma, USA) at 24 and 48 h post-infection. Subsequently the spleen and kidney of the fish were taken aseptically, and the recoveries of bacteria in the tissues were determined as previously [39]. The assay was performed in triplicate.

Transcriptional regulation of cadBA by CadC

In the study of cadC, we obtained cadC mutant TX01ΔcadC. The expression of cadBA operon in TX01 and TX01ΔcadC at acid condition was examined by RT-qPCR. To examine cadBA expression in in vitro conditions, the speculative promoter of cadBA (the 418 bp DNA upstream of cadBA operon), P418, was cloned with primers CadBAPF1/CadBAPR1 and was inserted into the BamHI site of pSC11, a promoter probe plasmid [41], which resulted in pSC418. pSC418 and pJR21C which expressed the CadC were introduced into E. coli DH5α by co-transformation, and cultured on X-gal (5-bromo-4-chloro-3-indolyl-beta-d-galactopyranoside) plate. Meanwhile, pJR21 was used as the control. To study the transcriptional regulation of P418 by CadC in vivo, the amplified P418 fragment was inserted into the SmaI site of p181-lux, a promoter probe plasmid based on pACYC177 and pGL28. The resulting plasmid was digested with SmiI and the fragment that included P418 and luciferase gene was inserted into the PmeI site of pJR21, resulting in pJR21-418-lx. pJR21-418-lx was introduced into TX01 and TX01ΔcadC which is the cadC mutant by electro-transformation, respectively. The transformants were screened on LB agar plates supplemented with 100 μg/mL polymyxin B and 20 μg/mL tetracycline. The log-phase transformants were cultured in normal conditions (pH = 7.0) or in LB broth (pH = 5.5) with 5 mM l-lysine for 1 h, and subjected to luciferase assay using the Firefly Luciferase Reporter Gene Assay Kit (Beyotime, China).

Statistical analysis

All data were analyzed statistically using the analysis of variance (ANOVA) with SPSS 18.0 software (SPSS Inc., Chicago, IL, USA), and P < 0.05 was considered statistically significant. Each experiment was performed three times, data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. Statistical significance (*, P < 0.05; **, P < 0.01) was obtained using an ANOVA test with SPSS 18.0 software (SPSS Inc., Chicago, IL, USA).

Results

Characterization of cadBA operon sequence and its co-transcription verification

Although some genes or factors were identified to contribute to acid resistance of E. tarda, there is not any report on the classical acid resistance systems in E. tarda. In order to further explore E. tarda’s mechanism of acid resistance, a lysine-dependent acid resistance (LDAR) system, CadBA, was identified and characterized in this study. The cadB of E. tarda (ETAE_0756) consists of 1332 bp ORF that encodes a lysine/cadaverine antiporter composed of 443 amino acids with a calculated molecular mass of 46.1 kDa and a theoretical pI of 9.01. Structural analysis suggested that CadB is a membrane protein with 12 transmembrane helices. Multiple sequence alignment shows that CadB shares high overall amino acid sequence identities (66–84%) with lysine/cadaverine antiporter homologues of Salmonella enterica, Klebsiella pneumoniae, E. coli, Shigella flexneri, Vibrio cholerae, and Aeromonas veronii (Additional file 1).

The cadA of E. tarda (ETAE_0757) consists of 2145 bp ORF that encodes a lysine decarboxylase composed of 714 amino acids with a calculated molecular mass of 81.0 kDa and a theoretical pI of 5.53. CadA includes an N-terminal domain (residues 14 to 124), a PLP-binding core domain (residues 131 to 441), and a C-terminal domain (residues 571 to 695), which contains a pyridoxal phosphate binding motif formed by 15 residues. Multiple sequence alignment showed that CadA shares high overall amino acid sequence identities (70–86%) with lysine decarboxylase homologues of E. coli, Salamae, A. veronii, V. cholerae, K. pneumoniae, and Shigella boydii (Additional file 1).

The cadB and cadA of numerous bacteria such as E. coli, V. cholerae, S. typhimurium are co-transcribed under one operon, providing advantages to counter low intracellular and extracellular pH. In E. tarda, there is a 148 bp intergenic spacer region between cadB and cadA (Figure 1A). To verify whether the two genes are co-transcribed, a pair of primers annealing to the 3′ end of cadB and 5′ end of cadA was designed. PCR was performed using total RNA, complementary DNA (cDNA), and genomic DNA (gDNA) of E. tarda as templates, respectively. The results show that the predicted PCR product of 795 bp was amplified in the reaction using cDNA and gDNA, but not in RNA (Figure 1B), which indicates that cadB and cadA are co-transcribed under one operon. The operon is named as cadBA. The upstream gene of the cadBA operon is a cadC encoded transcriptional regulator, and there is a predictive promoter between cadB and cadC, Pcad (Figure 1A).

Figure 1
figure 1

Schematic organization and co-transcriptional verification of the cadBA operon in E. tarda. A Schematic organization of the cadBA operon in E. tarda. The cadBA operon consists of cadB and cadA. The cadBA operon is under the control of the predictive promoter Pcad. The upstream gene of the cadBA operon is cadC, which encodes transcriptional regulator. Therefore, the operon was named cadBA. B Verification of cadB and cadA co-transcription. Genomic DNA and total RNA were isolated from overnight cultures of E. tarda. RNA was treated with DNase I and cDNA was synthesized. PCR were conducted with specific primer pair CadBAF/CadBAR using genomic DNA, cDNA, and RNA as the template. PCR products were analyzed by agarose gel electrophoresis.

Constructions of mutant strains TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA and complementary strains TX01ΔcadAC, and TX01ΔcadBC

To verify the function of the cadBA operon, the cadA and cadB genes were knocked out by markerless in-frame deletion of region encoding amino acid residues 173 to 513 and 113 to 324, respectively. Meanwhile, the whole operon was also knocked out by markerless in-frame deletion of the region from the N-terminal 113 amino acid residues of CadB to C-terminal 202 amino acid residues of CadA. Deletion of the genes were verified by PCR with specific primers and sequencing (Additional file 2). The resulting mutants were named TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA. The genetic complementation of TX01ΔcadA and TX01ΔcadB, TX01ΔcadAC and TX01ΔcadBC, were also obtained.

cadBA is involved in adversity resistance

In the process of infecting the host or host cell, pathogens inevitably face some environmental pressures, such as acid stress, iron deficiency, and oxidative stress. Previous studies indicated that cadBA operon in several bacteria were essential for physiological fitness to acid stress. We want to know whether cadBA in E. tarda has any effects on bacterial resistance to acid stress and some other environmental pressures. For this purpose, wild type strain TX01, cadBA operon deletion mutant strains TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA were cultured in LB broth with a pH range of 7.0 to 4.5. No difference was observed between the growth characteristics of the wild-type and three mutants under normal conditions (pH = 7.0) (Figure 2A). When exposed to the acidic environment (pH = 5.5 to 4.5), the growths of four strains were retarded to varying degrees. Compared to wild type strain TX01, three mutants were obviously retarded. TX01ΔcadA was most affected by acid stress, followed by TX01ΔcadBA and TX01ΔcadB, but the cell densities of three mutants were similar to that of TX01 at the stationary phase (Figures 2B–D). When the pH of the medium adjusted to pH 4.0, mutants failed to grow (data not shown).

Figure 2
figure 2

Growth analysis of E. tarda in different conditions. The wild-type TX01, its isogenic cadA, cadB and cadBA deletion mutant strains TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA were cultured to the exponential phase. After diluting serially, bacteria were cultured in fresh LB broth with pH adjusted to 7.0, 5.5, 5.0, and 4.5 (AD). Bacteria were added into fresh medium with 500 μM of diamide (E) and 50 μM of 2,2ʹ-dipyridyl (Dp) (F), respectively. Cell density was monitored at 2-h intervals by measuring the OD600. Experiment was performed three times, data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed.

When grown in LB medium containing 500 μM of oxidant diamide, the growths of four strains were retarded and three mutants displayed slightly slower growth rates than TX01 (Figure 2E). When grown in LB medium containing 50 μM of dipyridyl (a kind of iron chelator), the growths of four strains were retarded and three mutants displayed obviously slower growth rates than TX01, but there was no obvious difference of growth amongst three mutants (Figure 2F). These results suggest that the deletion of cadA or cadB does not impair the growth of E. tarda under normal conditions, but to a certain extent, affects bacterial growth under iron deficiency and oxidative stress, especially under acid stress.

cadBA participates in lysine metabolism

The inducible lysine decarboxylase can convert lysine to cadaverine, which plays an important role in pH homeostasis by neutralizing the acidic by-products of carbohydrate fermentation. In order to explore the mechanism that cadBA contributes to acid tolerance of E. tarda, we examined the effect of cadBA on activity of lysine decarboxylase. The LDC assay was performed and the result of qualitative measurement shows that TX01 exhibited observable purple after incubation in static culture for 10 h, which indicates an LDC positive phenotype (Figure 3A). However, three mutants exhibited faint yellow or light color, which indicates an LDC negative phenotype. After culturing for 20 h, TX01 exhibited a stronger purple and TX01ΔcadA and TX01ΔcadB displayed observable purple, but TX01ΔcadBA remained faint yellow. When culturing time reaches 30 h, all four strains displayed different degrees of purple. Complementary strains TX01ΔcadAC and TX01ΔcadBC could partially recover the positive phenotype of lysine decarboxylase (Figure 3A). The quantitative assays revealed that the lysine decarboxylase activity in the mutant TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA was 10.1, 26.2, 3.4 U, respectively, which all are significantly lower than that in TX01 (34.6 U). Among the three mutants, the lysine decarboxylase activity of TX01ΔcadBA was significantly lower than that of TX01ΔcadA, and the latter was significantly lower than that of TX01ΔcadB (Figure 3B). The lysine decarboxylase activity of complementary strain TX01ΔcadBC was similar to that of TX01, while that of complementary strain TX01ΔcadAC was dramatically higher than that of TX01ΔcadA but a little lower than that of TX01 (Figure 3B).

Figure 3
figure 3

The activities of lysine decarboxylase in E. tarda. A TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC were cultured in lysine decarboxylase broth with lysine in static condition at 28 ℃ for 10, 20 and 30 h, respectively. A light purple indicates a weak positive result. Purple and yellow indicate the presence and absence of LDC, respectively. B The same strains were cultured in LB broth to an OD600 of 0.5 and normalized to an OD600 of 1.0, buffered at pH 6.8. The activities of lysine decarboxylase were determined as described in the text. The experiment was performed three times, data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01.

cadBA mutations enhance bacterial biofilm formation, notably under acid conditions

Bacterial growth in biofilm mode enhances microbial survival and safeguards them from environmental stresses, including acid stress. We want to know whether the defect in acid tolerance caused by the deletion of cadBA affects bacterial biofilm growth, so the biofilm formations of TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC were determined by CV staining. As shown in Figure 4A, the biofilm forming capabilities of TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA were significantly stronger than that of TX01 when bacteria were cultured in normal LB medium (pH = 7.0) for 24 h. Consistently, images of the biofilms of four strains obtained by confocal laser scanning microscopy (CLSM) also showed that thicknesses and densities of the biofilm of three mutants were stronger than the wild type, and thickness and density of TX01ΔcadB was the strongest amongst three mutants (Figure 4B). When bacteria were cultured in acid LB medium (pH = 5.5), the biofilm growths of all the strains exhibited a significant increase, especially TX01ΔcadBA, compared to those in normal medium (Figure 4A). The complementary strains TX01ΔcadAC and TX01ΔcadBC restored the capacity of biofilm formation of TX01ΔcadA, and TX01ΔcadB, respectively. These results suggest that cadBA negatively participates in biofilm formation, especially under acid conditions.

Figure 4
figure 4

Effects of cadBA mutations on biofilm formation. A Biofilm-forming capacity of E. tarda in normal and acid conditions. TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC were inoculated into LB broth at pH = 7.0 and 5.5, then incubated in polystyrene plates for 24 h. Biofilm formations were determined by measuring the A570 of the final eluates of crystal violet staining. Data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01; *, P < 0.05. B The viability of biofilm growth of E.tarda was determined by confocal laser scanning microscopy (CLSM). Cells in the biofilms were stained with a BacLight LIVE/DEAD kit to reveal viable (green fluorescence) and non-viable (red fluorescence) bacteria.

cadBA mutations impair bacterial motility under acid conditions

To investigate whether deletion of cadBA has any effect on bacterial motility, six strains were inoculated into swimming plates and bacterial motilities were observed for 18 h. The swimming assay shows that on the neutral LB plate, TX01 and three mutants exhibited similar and little swimming zones (Figure 5A). However, in the acid swimming plate except TX01ΔcadA, motility remained weak, other three strains’ moveability were obviously enhanced (Figure 5B), but the swimming zones of TX01ΔcadB and TX01ΔcadBA were significantly shorter than that of TX01 (Figure 5C). The swimming zone of complementary strain TX01ΔcadBC was comparative to that of TX01, while the swimming zone of complementary strain TX01ΔcadAC was shorter than that of TX01 (Additional file 3), which indicates TX01ΔcadAC could have partially recovered swimming ability in the acid swimming plate. These results suggest that cadBA operon mutations impair E. tarda's motility under acid conditions.

Figure 5
figure 5

Effects of cadBA mutations on motility of E. tarda. TX01, TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA were cultured in LB medium to an OD600 of 0.5, then aliquots of cell suspensions (1 μL) were inoculated into the center of swimming plates including 0.3% (W/V) agar with pH = 7.0 (A) or pH = 5.5 (B) at 28 °C for 18 h. C, The diameter of the swimming zone from the swimming plate at pH = 5.5. Data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01; *, P < 0.05.

cadBA deletions reduce the ability of adhesion and invasion to HeLa cells

The above results show that the deficiency of cadBA increased E. tarda’s biofilm formation but decreased motility in an acid environment, and reduced adversity resistance. These physiological phenotypes are all related to bacterial pathogenicity, so we want to know whether the deletion of cadBA could affect the virulence of E. tarda. To evaluate the participation of cadBA operon in the virulence of E. tarda, HeLa cells were infected with the same dose of TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC strains for 1 and 2 h, then were washed three times and lysed. The bacteria adhering to the surface of cells and invading into the interior were examined. The results of plate counting indicate that the amounts of TX01ΔcadA, TX01ΔcadB, and TX01ΔcadBA from HeLa cells were remarkably less than that of TX01 at 1 and 2 h post-infection, and the amounts of TX01ΔcadA and TX01ΔcadBA from HeLa cells were also significantly less than that of TX01ΔcadB at both time points (Figure 6A). The amounts of complementary strain TX01ΔcadAC from HeLa cells were comparative to those of TX01. However, the amount of complementary strain TX01ΔcadBC from HeLa cells was more than that of TX01 at 1 h post-infection, but the amounts of both were comparative at 2 h post-infection. These observations suggest that the mutations of cadBA impair the adhesion and invasion of E. tarda to non-phagocytes.

Figure 6
figure 6

Effects of cadBA mutations on cellular infection and replication. A Invasion of HeLa cells by E. tarda. HeLa cells were infected with the same dose of E. tarda TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC and TX01ΔcadBC strains for 1 and 2 h, and washed with PBS. Then, HeLa cells were lysed and the CFU were counted. B Replication of E. tarda in macrophages. The murine macrophage cell line RAW264.7 was infected with E. tarda and mutants above mentioned for 2 h, followed by treatment with gentamicin for 2 h to kill extracellular bacteria. After being washed with PBS, the cells were incubated for the time intervals indicated. Then, the cells were lysed and the CFU were counted. Data are the means of three independent experiments and presented as means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01; *, P < 0.05. C, TX01, TX01ΔcadA, TX01ΔcadB and TX01ΔcadBA containing pGFPuv plasmid were used to infect RAW264.7 cells for 0, 2 and 4 h. DNA was stained blue by DAPI, the cells were observed by confocal microscopy.

cadBA is required for bacterial survival and replication in macrophages

E. tarda prefers an intracellular lifestyle in host cells to escape the innate immunity and to multiply. Next, we examined the role of cadBA in phagocyte survival and replication of E. tarda. The same dose of TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC were used to infect murine macrophage cell line RAW264.7. After eliminating extracellular bacteria, RAW264.7 cells were cultured for different hours and were lysed to determine the intracellular bacteria. From the results of plate counting, we found that the amounts of TX01ΔcadA, TX01ΔcadB and TX01ΔcadBA from the intracellular macrophage cells were remarkably less than those of TX01 at 0, 2, 4, and 8 h post-infection, and the amount of TX01ΔcadBA was basically the least amongst the three mutants (Figure 6B). The amounts of complementary strains TX01ΔcadAC and TX01ΔcadBC from the intracellular macrophage cells were similar to those of TX01.

To observe the living bacteria in macrophages, E. tarda containing pGFPuv plasmid was used to infect RAW264.7 cells. By means of confocal microscopy, it was clearly observed that the fluorescence intensities from TX01ΔcadA-, TX01ΔcadB-, and TX01ΔcadBA-infected macrophages were weaker than those from TX01-infected cells at 0, 2 and 4 h post-infection (Figure 6C). These results suggest that the mutations of cadBA impair the survival and replication of E. tarda in host phagocytes.

cadBA is a requisite for bacterial dissemination in host tissues

An in vitro experiment has confirmed the participation of cadBA operon in E. tarda’s invasion of host cells. Next, in vivo experiments were performed to determine the contribution of cadBA operon to virulence of E. tarda. To do this, the same doses of strains (TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC) were used to infect tilapia. Viable bacteria from spleen and kidney were determined by plate counting at 24 and 48 h post-infection. The results show that bacterial amounts in both tissues from TX01ΔcadA-, TX01ΔcadB- and TX01ΔcadBA-infected tilapias were obviously less than those from TX01-infected tilapias at both infection time points. And the amounts of two complementary strains TX01ΔcadAC and TX01ΔcadBC from infected tilapias were basically equal to those of TX01 (Figure 7). These results imply that the cadBA operon is involved in the dissemination of E. tarda in the fish tissue.

Figure 7
figure 7

Bacterial dissemination in the fish tissues. Tilapia were infected with the same dose of TX01, TX01ΔcadA, TX01ΔcadB, TX01ΔcadBA, TX01ΔcadAC, and TX01ΔcadBC. The recoveries of bacteria in spleen (A) and kidney (B) were determined by plate counting at 24 and 48 h post-infection. Data are presented as means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01.

cadBA expression is up-regulated upon acid stress

Since cadBA was closely related to acid tolerance, we wanted to survey the expression of cadBA under acid pressure. E. tarda in exponential phase were subjected to an acidic medium (pH 5.5) for 1 h, then RNA was extracted and cDNA was obtained. RT-qPCR shows that the expression of cadB, cadA, and cadBA was up-regulated 685.5-, 1808.7-, and 1204.1-folds respectively, upon acid stimulation, which was significantly higher than that of the control (Figure 8). The immediate upstream of cadBA is cadC, so we also surveyed its expression and found that cadC expression was significantly enhanced (3.2-folds) by acid stimulation (Figure 8). These results indicate that the cadBA operon is closely involved in acid tolerance of E. tarda.

Figure 8
figure 8

The expression of acid resistance genes from TX01 under acidic conditions. The exponential phase of overnight cultures of TX01 were grown in normal LB medium at pH 7.0 and acidic medium at pH 5.5 for 1 h. The relative expression of cadB, cadA, cadBA, and cadC were determined by RT-qPCR. The fold difference derived from the values under acidic conditions compared with the values under normal conditions. Data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01; *, P < 0.05.

cadBA expression is regulated by CadC

Since cadC is located upstream of the cadBA operon and defined as a transcriptional regulator, and the expression of cadC was induced by acid stimulation, we speculated that the expression of cadBA is regulated by CadC. In another study, we obtained a cadC mutant (named TX01ΔcadC). We first investigated the expression of cadBA in wild strain TX01 and TX01ΔcadC under normal conditions and acidic conditions. RT-qPCR shows that under normal conditions, the expression of cadA, cadB, and cadBA in TX01ΔcadC was significantly lower than that in TX01, the values were 2.6-, 1.8-, and 1.6-fold, respectively. Under acidic conditions, the expression of cadA, cadB, and cadBA in TX01ΔcadC was down regulated by 2382.0-, 2848.0-, and 13,777.6-folds respectively, compared to TX01 (Figure 9A). These results support the conclusion that CadC is an activator of cadBA transcription, and its regulator role is mainly displayed under acidic conditions.

Figure 9
figure 9

Transcriptional regulation of cadBA by CadC in vitro or in vivo. A Expression profiles of cadBA operon in normal and acidic conditions. E. tarda TX01 and TX01ΔcadC in logarithmic growth phase was culture in normal LB (pH = 7.0) or in acidic LB (pH = 5.5) for 1 h, then the expression of cadB, cadA, and cadBA were examined by RT-qPCR. For convenience of comparison, the expression level of cadBA operon in wild type TX01 set as 1. B The promoter activity of cadBA operon was regulated by CadC in vitro. DH5α/pSC418/pJR21 and DH5α/pSC418/pJR21C were streaked and cultured on an X-gal plate at pH 5.5 in the presence of 5 mM L-lysin. The depth of blue indicates the strength of promoter activity. C The promoter activity of cadBA operon was regulated by CadC in vivo. The wild-type TX01 and TX01ΔcadC mutant carrying the reporter plasmid pJR21-418-lx were cultured to the exponential phase. Then strains were transferred acid LB media (pH = 5.5) supplemented with 5 mM l-lysine and grown for 1 h. The transcriptional regulation of promoter of cadBA was assessed by measuring luciferase activity. Data are the means of three independent experiments and presented as means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01; *, P < 0.05.

To detect the regulatory mechanism of CadC on cadBA, the speculative promoter of cadBA, P418, was cloned to a promoter probe plasmid pSC11 resulting in DH5α/pSC418. Meanwhile, the expression plasmid of cadC was constructed and named pJR21C. DH5α/pSC418 cultured on X-gal plate at pH 7.0 and 5.5 in the presence of 5 mM l-lysine shows a weak blue phenotype, indicating that the P418 promoter has a weak promoter activity. When pJR21C was transformed into DH5α/pSC418, the blue color of the recombinant strain (DH5α/pSC418/pJR21C) on the X-gal plate at pH 7.0 remained unchanged in the presence of 5 mM l-lysine, compared to that of the control (DH5α/pSC418/pJR21) (data not shown). However, the blue color of DH5α/pSC418/pJR21C on X-gal plate at pH 5.5 in the presence of 5 mM l-lysine was obviously enhanced, compared to that of the control (Figure 9B). These results suggest that CadC positively regulates the promoter activity of cadBA under acidic conditions.

To further analyze the regulation of CadC on cadBA in vivo, promoter reporter plasmid pJR21-418-lx was introduced into TX01 and TX01△cadC, and the luminescence induced by promoter P418 was detected. The results show the relative light units of (RLU) of TX01 were 463.5 U and 8513.58 U under normal conditions and acidic induced conditions, respectively, which both are significantly higher than that in TX01△cadC (213.7 U and 757.95 U, respectively), especially under acidic induction conditions (Figure 9C). These results indicate that CadC positively regulates the transcription of cadBA in E. tarda, especially under acidic conditions.

Discussion

Pathogenic and commensal bacteria often encounter a lot of strong and mild acidic environments both inside and outside hosts, for example, acid soil and food, the gastrointestinal tract, dental plaque and macrophage phagosome [14]. In general, bacteria are able to maintain a fairly constant internal pH for survival, which is due to evolving acid tolerance response (ATR) and acid resistance (AR) protection mechanisms [

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

E. tarda :

Edwardsiella tarda

E. anguillarum :

Edwardsiella anguillarum

E. ictaluri :

Edwardsiella ictaluri

E. hoshinae :

Edwardsiella hoshinae

E. piscicida :

Edwardsiella piscicida

E. coli :

Escherichia coli

V. cholera :

Vibrio cholera

S. typhimurium :

Salmonella typhimurium

V. vulnificus :

Vibrio vulnificus

AR:

acid-resistance

ATR:

acid tolerance response

PLP:

phosphopyridoxal

GDAR:

glutamic acid-dependent acid resistance

ADAR:

arginine-dependent acid resistance

LDAR:

lysine-dependent acid resistance

ODAR:

ornithine-dependent acid resistance

LB:

Luria–Bertani broth

PCR:

Polymerase Chain Reaction

RT-PCR:

reverse transcription-PCR

RT-qPCR:

quantitative real-time reverse transcription-PCR

ORF:

open reading frames

CFU:

colony forming unit

Dp:

2,2ʹ-Dipyridyl

LDC:

lysine decarboxylase

OD:

optical density

A :

absorption

CV:

crystal violet

CLSM:

confocal laser scanning microscopy

PBS:

phosphate-buffered saline

DMEM:

Dulbecco’s Modified Eagle’s medium

MOI:

multiplicity of infection

DAPI:

4ʹ, 6-Diamidino-2-phenylindole

X-gal:

5-Bromo-4-chloro-3-indolyl-beta-d-galactopyranoside

U:

unite

TM:

transmembrane domains

i.m.:

intramuscular

MS222:

tricaine methanesulfonate

References

  1. Ewing WH, Mcwhorter AC, Escobar MR, Lubin AH (1965) Edwardsiella, a new genus of Enterobacteriaceae based on a new species, E. tarda. Int J Syst Evol Microbiol 15:33–38

    Google Scholar 

  2. Abayneh T, Colquhoun DJ, Sørum H (2013) Edwardsiella piscicida sp. nov., a novel species pathogenic to fish. J Appl Microbiol 114:644–654

    Article  CAS  PubMed  Google Scholar 

  3. Leung KY, Wang Q, Yang Z, Siame BA (2019) Edwardsiella piscicida: a versatile emerging pathogen of fish. Virulence 10:555–567

    Article  PubMed  PubMed Central  Google Scholar 

  4. Griffin MJ, Quiniou SM, Cody T, Tabuchi M, Ware C, Cipriano RC, Mauel MJ, Soto E (2013) Comparative analysis of Edwardsiella isolates from fish in the eastern United States identifies two distinct genetic taxa amongst organisms phenotypically classified as E. tarda. Vet Microbiol 165:358–372

    Article  CAS  PubMed  Google Scholar 

  5. Wang C, Dong XS, Yang YY, Xu GJ, Wu MM, Yan FJ, Zhang LG, An L, Fu PS, Wang XR, Su YB, Meng QL (2021) Metabolites in the TCA cycle promote resistance to chloramphenicol of Edwardsiella tarda. J Proteome Res 20:972–981

    Article  CAS  PubMed  Google Scholar 

  6. Han HH, Li T, Wang ZL, Teng D, Mao RY, Hao Y, Yang N, Wang XM, Wang JH (2020) Improved stability and activity of a marine peptide-N6NH2 against Edwardsiella tarda and its preliminary application in fish. Mar Drugs 18:650

    Article  CAS  PubMed Central  Google Scholar 

  7. Sayed M, Ozdemir O, Essa M, Olivier A, Karsi A, Lawrence ML, Abdelhamed H (2021) Virulence and live vaccine potential of Edwardsiella piscicida phoP and phoQ mutants in catfish against edwardsiellosis. J Fish Dis 44:1463–1474

    Article  CAS  PubMed  Google Scholar 

  8. Buján N, Toranzo AE, Magariños B (2018) Edwardsiella piscicida: a significant bacterial pathogen of cultured fish. Dis Aquat Organ 131:59–71

    Article  PubMed  CAS  Google Scholar 

  9. Bockemühl J, Pan-Urai R, Burkhardt F (1971) Edwardsiella tarda associated with human disease. Pathol Microbiol (Basel) 37:393–401

    Google Scholar 

  10. Lee W, Oh JY, Kim EA, Kang N, Kim KN, Ahn G, Jeon YJ (2016) A prebiotic role of Ecklonia cava improves the mortality of Edwardsiella tarda-infected zebrafish models via regulating the growth of lactic acid bacteria and pathogen bacteria. Fish Shellfish Immunol 54:620–628

    Article  CAS  PubMed  Google Scholar 

  11. Gao YL, Tang XQ, Sheng XZ, **ng J, Zhan WB (2015) Immune responses of flounder Paralichthys olivaceus vaccinated by immersion of formalin-inactivated Edwardsiella tarda following hyperosmotic treatment. Dis Aquat Organ 116:111–120

    Article  CAS  PubMed  Google Scholar 

  12. Wang IK, Kuo HL, Chen YM, Lin CL, Chang HY, Chuang FR, Lee MH (2005) Extraintestinal manifestations of Edwardsiella tarda infection. Int J Clin Pract 59:917–921

    Article  PubMed  Google Scholar 

  13. Nelson JJ, Nelson CA, Carter JE (2009) Extraintestinal manifestations of Edwardsiella tarda infection: a 10-year retrospective review. J La State Med Soc 161:103–106

    PubMed  Google Scholar 

  14. Lund P, Tramonti A, De Biase D (2014) Co** with low pH: molecular strategies in neutralophilic bacteria. FEMS Microbiol Rev 38:1091–1125

    Article  CAS  PubMed  Google Scholar 

  15. Park YK, Bearson B, Bang SH, Bang IS, Foster JW (1996) Internal pH crisis, lysine decarboxylase and the acid tolerance response of Salmonella typhimurium. Mol Microbiol 20:605–611

    Article  CAS  PubMed  Google Scholar 

  16. Brameyer S, Hoyer E, Bibinger S, Burdack K, Lassak J, Jung K (2020) Molecular design of a signaling system influences noise in protein abundance under acid stress in different γ-Proteobacteria. J Bacteriol. https://doi.org/10.1128/JB.00121-20

    Article  PubMed  PubMed Central  Google Scholar 

  17. Xu Y, Zhao Z, Tong W, Ding Y, Liu B, Shi Y, Wang J, Sun S, Liu M, Wang Y, Qi Q, **an M, Zhao G (2020) An acid-tolerance response system protecting exponentially growing Escherichia coli. Nat Commun 11:1496

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Krammer EM, Prevost M (2019) Function and regulation of acid resistance antiporters. J Membr Biol 252:465–481

    Article  CAS  PubMed  Google Scholar 

  19. De Biase D, Tramonti A, Bossa F, Visca P (1999) The response to stationary-phase stress conditions in Escherichia coli: role and regulation of the glutamic acid decarboxylase system. Mol Microbiol 32:1198–1211

    Article  PubMed  Google Scholar 

  20. Iyer R, Williams C, Miller C (2003) Arginine-agmatine antiporter in extreme acid resistance in Escherichia coli. J Bacteriol 185:6556–6561

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Soksawatmaekhin W, Kuraishi A, Sakata K, Kashiwagi K, Igarashi K (2004) Excretion and uptake of cadaverine by CadB and its physiological functions in Escherichia coli. Mol Microbiol 51:1401–1412

    Article  CAS  PubMed  Google Scholar 

  22. Kashiwagi K, Shibuya S, Tomitori H, Kuraishi A, Igarashi K (1997) Excretion and uptake of putrescine by the PotE protein in Escherichia coli. J Biol Chem 272:6318–6323

    Article  CAS  PubMed  Google Scholar 

  23. Kanjee U, Gutsche I, Alexopoulos E, Zhao B, Bakkouri EI, M, Thibault G, Liu K, Ramachandran S, Snider J, Pai EF, Houry WA, (2011) Linkage between the bacterial acid stress and stringent responses: the structure of the inducible lysine decarboxylase. Embo J 30:931–944

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Diez-Gonzalez F, Karaibrahimoglu Y (2004) Comparison of the glutamate-, arginine- and lysine-dependent acid resistance systems in Escherichia coli O157:H7. J Appl Microbiol 96:1237–1244

    Article  CAS  PubMed  Google Scholar 

  25. Auger EA, Redding KE, Plumb T, Childs LC, Meng SY, Bennett GN (1989) Construction of lac fusions to the inducible arginine- and lysine decarboxylase genes of Escherichia coli K12. Mol Microbiol 3:609–620

    Article  CAS  PubMed  Google Scholar 

  26. Sabo DL, Boeker EA, Byers B, Waron H, Fischer EH (1974) Purification and physical properties of inducible Escherichia coli lysine decarboxylase. Biochemistry 13:662–670

    Article  CAS  PubMed  Google Scholar 

  27. Dell CL, Neely MN, Olson ER (1994) Altered pH and lysine signalling mutants of cadC, a gene encoding a membrane-bound transcriptional activator of the Escherichia coli cadBA operon. Mol Microbiol 14:7–16

    Article  CAS  PubMed  Google Scholar 

  28. Miller VL, Taylor RK, Mekalanos JJ (1987) Cholera toxin transcriptional activator toxR is a transmembrane DNA binding protein. Cell 48:271–279

    Article  PubMed  Google Scholar 

  29. Rauschmeier M, Schüppel V, Tetsch L, Jung K (2014) New insights into the interplay between the lysine transporter LysP and the pH sensor CadC in Escherichia coli. J Mol Biol 426:215–229

    Article  CAS  PubMed  Google Scholar 

  30. Schlundt A, Buchner S, Janowski R, Heydenreich T, Heermann R, Lassak J, Geerlof A, Stehle R, Niessing D, Jung K, Sattler M (2017) Structure-function analysis of the DNA-binding domain of a transmembrane transcriptional activator. Sci Rep 7:1051

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Meng SY, Bennett GN (1992) Nucleotide sequence of the Escherichia coli cad operon: a system for neutralization of low extracellular pH. J Bacteriol 174:2659–2669

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rhee JE, Kim KS, Choi SH (2005) CadC activates pH-dependent expression of the Vibrio vulnificus cadBA operon at a distance through direct binding to an upstream region. J Bacteriol 187:7870–7875

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang M, Jiao XD, Hu YH, Sun L (2009) Attenuation of Edwardsiella tarda virulence by small peptides that interfere with LuxS/autoinducer type 2 quorum sensing. Appl Environ Microbiol 75:3882–3890

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ma DM, Gu HJ, Shi YJ, Huang HQ, Sun DM, Hu YH (2021) Edwardsiella piscicida YefM-YoeB: a type II Toxin-Antitoxin system that is related to antibiotic resistance, biofilm formation, serum survival, and host infection. Front Microbiol 12:646299

    Article  PubMed  PubMed Central  Google Scholar 

  35. Hu YH, Sun L (2016) The global regulatory effect of Edwardsiella tarda Fur on iron acquisition, stress resistance, and host infection: a proteomics-based interpretation. J Proteomics 140:100–110

    Article  CAS  PubMed  Google Scholar 

  36. Ante VM, Bina XR, Bina JE (2015) The LysR-type regulator LeuO regulates the acid tolerance response in Vibrio cholerae. Microbiology 161:2434–2443

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shi YJ, Fang QJ, Huang HQ, Gong CG, Hu YH (2019) HutZ is required for biofilm formation and contributes to the pathogenicity of Edwardsiella piscicida. Vet Res 50:76

    Article  PubMed  PubMed Central  Google Scholar 

  38. Du HH, Zhou HZ, Tang P, Huang HQ, Liu M, Hu YH (2018) Global discovery of small RNAs in the fish pathogen Edwardsiella piscicida: key regulator of adversity and pathogenicity. Vet Res 49:120

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. **e JH, Zhao QY, Huang HQ, Fang ZG, Hu YH (2021) Edwardsiella piscicida HigB: a type II toxin that is essential to oxidative resistance, biofilm formation, serum survival, intracellular propagation, and host infection. J Aquaculture 535:736382

    Article  CAS  Google Scholar 

  40. Wang D, Gong CG, Gu HJ, Huang HQ, **an JN, Hu YH (2021) Bicistronic operon YhaO-YhaM contributes to antibiotic resistance and virulence of pathogen Edwardsiella piscicida. Aquaculture 541:736849

    Article  CAS  Google Scholar 

  41. Wang F, Cheng S, Sun K, Sun L (2008) Molecular analysis of the fur (ferric uptake regulator) gene of a pathogenic Edwardsiella tarda strain. J Microbiol 46:350–355

    Article  CAS  PubMed  Google Scholar 

  42. Perez JC, Groisman EA (2007) Acid pH activation of the PmrA/PmrB two-component regulatory system of Salmonella enterica. Mol Microbiol 63:283–293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jessop M, Liesche C, Felix J, Desfosses A, Baulard M, Adam V, Fraudeau A, Huard K, Effantin G, Kleman JP, Bacia-Verloop M, Bourgeois D, Gutsche I (2021) Supramolecular assembly of the Escherichia coli LdcI upon acid stress. Proc Natl Acad Sci U S A 118:e2014383118

    Article  CAS  PubMed  Google Scholar 

  44. Merrell DS, Camilli A (2000) Regulation of vibrio cholerae genes required for acid tolerance by a member of the “ToxR-like” family of transcriptional regulators. J Bacteriol 182:5342–5350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lee YH, Kim BH, Kim JH, Yoon WS, Bang SH, Park YK (2007) CadC has a global translational effect during acid adaptation in Salmonella enterica serovar Typhimurium. J Bacteriol 189:2417–2425

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ante VM, Bina XR, Bina JE (2015) The LysR-type regulator LeuO regulates the acid tolerance response in Vibrio cholerae. Microbiology (Reading) 161:2434–2443

    Article  CAS  Google Scholar 

  47. Fang QJ, Han YX, Shi YJ, Huang HQ, Fang ZG, Hu YH (2019) Universal stress proteins contribute Edwardsiella piscicida adversity resistance and pathogenicity and promote blocking host immune response. Fish Shellfish Immunol 95:248–258

    Article  CAS  PubMed  Google Scholar 

  48. Li MF, Jia BB, Sun YY, Sun L (2020) The translocation and assembly module (TAM) of Edwardsiella tarda is essential for stress resistance and host infection. Front Microbiol 11:1743

    Article  PubMed  PubMed Central  Google Scholar 

  49. Wang BY, Huang HQ, Li S, Tang P, Dai HF, **an JA, Sun DM, Hu YH (2019) Thioredoxin H (TrxH) contributes to adversity adaptation and pathogenicity of Edwardsiella piscicida. Vet Res 50:26

    Article  PubMed  PubMed Central  Google Scholar 

  50. Liu E, Ye J, Song SS, Wang K, Zhang YX, Zhang HZ (2014) Impact of co-deficiency of RpoN and RpoS on stress tolerance, virulence and gene regulation in Edwardsiella tarda. J Basic Microbiol 54:678–687

    Article  CAS  PubMed  Google Scholar 

  51. Tanaka Y, Kimura B, Takahashi H, Watanabe T, Obata H, Kai A, Morozumi S, Fujii T (2008) Lysine decarboxylase of Vibrio parahaemolyticus: kinetics of transcription and role in acid resistance. J Appl Microbiol 104:1283–1293

    Article  CAS  PubMed  Google Scholar 

  52. Xu H, Lee HY, Ahn J (2008) Cross-protective effect of acid-adapted Salmonella enterica on resistance to lethal acid and cold stress conditions. Lett Appl Microbiol 47:290–297

    Article  CAS  PubMed  Google Scholar 

  53. Samartzidou H, Mehrazin M, Xu Z, Benedik MJ, Delcour AH (2003) Cadaverine inhibition of porin plays a role in cell survival at acidic pH. J Bacteriol 185:13–19

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kojima S, Kaneko J, Abe N, Takatsuka Y, Kamio Y (2011) Cadaverine covalently linked to the peptidoglycan serves as the correct constituent for the anchoring mechanism between the outer membrane and peptidoglycan in Selenomonas ruminantium. J Bacteriol 193:2347–2350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kamio Y, Pösö H, Terawaki Y, Paulin L (1986) Cadaverine covalently linked to a peptidoglycan is an essential constituent of the peptidoglycan necessary for the normal growth in Selenomonas ruminantium. J Biol Chem 261:6585–6589

    Article  CAS  PubMed  Google Scholar 

  56. Hirao T, Sato M, Shirahata A, Kamio Y (2000) Covalent linkage of polyamines to peptidoglycan in Anaerovibrio lipolytica. J Bacteriol 182:1154–1157

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hamana K, Saito T, Okada M, Sakamoto A, Hosoya R (2002) Covalently linked polyamines in the cell wall peptidoglycan of Selenomonas, Anaeromusa, Dendrosporobacter, Acidaminococcus and Anaerovibrio belonging to the Sporomusa subbranch. J Gen Appl Microbiol 48:177–180

    Article  CAS  PubMed  Google Scholar 

  58. Han LF, Yuan JJ, Ao XL, Lin SJ, Han X, Ye HH (2018) Biochemical characterization and phylogenetic analysis of the virulence factor lysine decarboxylase from Vibrio vulnificus. Front Microbiol 9:3082

    Article  PubMed  PubMed Central  Google Scholar 

  59. Kikuchi Y, Kojima H, Tanaka T, Takatsuka Y, Kamio Y (1997) Characterization of a second lysine decarboxylase isolated from Escherichia coli. J Bacteriol 179:4486–4492

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Merrell DS, Camilli A (1999) The cadA gene of Vibrio cholerae is induced during infection and plays a role in acid tolerance. Mol Microbiol 34:836–849

    Article  CAS  PubMed  Google Scholar 

  61. Wang D, Xu AM, Elmerich C, Ma LZ (2017) Biofilm formation enables free-living nitrogen-fixing rhizobacteria to fix nitrogen under aerobic conditions. ISME J 11:1602–1613

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Li Q, Sand W (2017) Mechanical and chemical studies on EPS from Sulfobacillus thermosulfidooxidans: from planktonic to biofilm cells. Colloids Surf B Biointerfaces 153:34–40

    Article  CAS  PubMed  Google Scholar 

  63. Hu WB, Feng SS, Tong YJ, Zhang HL, Yang HL (2020) Adaptive defensive mechanism of bioleaching microorganisms under extremely environmental acid stress: advances and perspectives. Biotechnol Adv 42:107580

    Article  CAS  PubMed  Google Scholar 

  64. Edwards SJ, Kjellerup BV (2013) Applications of biofilms in bioremediation and biotransformation of persistent organic pollutants, pharmaceuticals/personal care products, and heavy metals. Appl Microbiol Biotechnol 97:9909–9921

    Article  CAS  PubMed  Google Scholar 

  65. Boisen G, Davies JR, Neilands J (2021) Acid tolerance in early colonizers of oral biofilms. BMC Microbiol 21:45

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Williams P, Cámara M (2009) Quorum sensing and environmental adaptation in Pseudomonas aeruginosa: a tale of regulatory networks and multifunctional signal molecules. Curr Opin Microbiol 12:182–191

    Article  CAS  PubMed  Google Scholar 

  67. Kim K, Kim JN, Lim BS, Ahn SJ (2021) Urethane dimethacrylate influences the cariogenic properties of Streptococcus mutans. Materials (Basel) 14:1015

    Article  CAS  Google Scholar 

  68. Vazquez-Juarez RC, Kuriakose JA, Rasko DA, Ritchie JM, Kendall MM, Slater TM, Sinha M, Luxon BA, Popov VL, Waldor MK, Sperandio V, Torres AG (2008) CadA negatively regulates Escherichia coli O157:H7 adherence and intestinal colonization. Infect Immun 76:5072–5081

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Jores J, Torres AG, Wagner S, Tutt CB, Kaper JB, Wieler LH (2006) Identification and characterization of “pathoadaptive mutations” of the cadBA operon in several intestinal Escherichia coli. Int J Med Microbiol 296:547–552

    Article  CAS  PubMed  Google Scholar 

  70. Maurelli AT, Fernández RE, Bloch CA, Rode CK, Fasano A (1998) “Black holes” and bacterial pathogenicity: a large genomic deletion that enhances the virulence of Shigella spp. and enteroinvasive Escherichia coli. Proc Natl Acad Sci U S A 95:3943–3948

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nakamya MF, Ayoola MB, Park S, Shack LA, Swiatlo E, Nanduri B (2018) The role of cadaverine synthesis on pneumococcal capsule and protein expression. Med Sci (Basel) 6:8

    Google Scholar 

  72. Tao LT, Kang YH, Zhang L, Shi QM, Li Y, Wu TL, Qian AD, Sun WW, Shan XF (2020) Cadaverine reverse transporter (CadB protein) contributes to the virulence of Aeromonas veronii TH0426. Int Microbiol 23:489–499

    Article  CAS  PubMed  Google Scholar 

  73. Kuper C, Jung K (2005) CadC-mediated activation of the cadBA promoter in Escherichia coli. J Mol Microbiol Biotechnol 10:26–39

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to thank professor Jianqiang Lin at Shandong University for his kind provision of plamids; Associate professor Hao Long at Hainan University for providing reagents; Professor Huiqin Huang at Institute of Tropical Bioscience and Biotechnology for technical assistances.

Funding

This work was supported by the Hainan Provincial Natural Science Foundation of China (2019CXTD413), the Central Public-interest Scientific Institution Basal Research Fund for Chinese Academy of Tropical Agricultural Sciences (1630052019003, 1630052021020), National Natural Science Foundation of China (41476138, 21777060), the Basic Scientific Research Project of the Higher Education Institutions of Heilongjiang Province (2020-KYYWF-0234), Joint Guidance Project of Natural Science Foundation of Heilongjiang Province (LH2019H058).

Author information

Authors and Affiliations

Authors

Contributions

CMD performed the experiments, analyzed the data, and wrote the paper. XPH participated in the infection experiments, HJG participated in the expression analysis. DMW and YHH designed the experiment. YHH wrote the paper. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Dongmei Wu or Yonghua Hu.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the ethics committee of Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences (ID Number: ITBB202001). Efforts were taken to ensure that all research animals received good care and humane treatment.

Competing interests

The authors have no conflicting commercial or financial interest in publishing this paper.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1.

Sequence alignment of CadA (A) and CadB (B) homologues. Gaps used to maximize the alignment are indicated by dots. The complete conserved amino acid residues are shown in dark blue. The amino acid residues with conservative degree higher than 75% are shown in pink. The black line indicates the N-terminal domain. The green and orange lines indicate the PLP-binding core domain and C-terminal domain, respectively. Red triangles indicate PLP binding sites. Conserve PLP binding motif is yellow boxed. The blue line indicates transmembrane (TM) domains. The GenBank accession numbers of CadA homologues are as follows: Edwardsiella tarda, ACY83602.1; Escherichia coli str. K-12 substr, NP_418555.1; Salamae, VEA01344.1; Aeromonas veronii, QHC07268.1; Vibrio cholerae, WP_188372281.1; Klebsiella pneumoniae, AZH95435.1; Shigella boydii, PHU85906.1. The GenBank accession numbers of CadB homologues are as follows: E. tarda, ACY83601.1; Salmonella enterica, WP_000100010.1; Klebsiella pneumoniae, WP_110924908.1; Escherichia coli, WP_175119720.1; Shigella flexneri, NP_709997.1; Vibrio cholerae, WP_189017881.1; Aeromonas veronii, WP_118881724.1.

Additional file 2.

Validation of TX01 mutants by PCR amplification. Lanes 1 and 7 were the 661-bp (deletion of cadA allele) fragments amplified from TX01ΔcadA and TX01ΔcadBA genomic DNA with primers CadAKOF3/CadAKOR3, respectively. Lane 4 was 1684-bp cadA fragment. Lanes 5 and 8 were the 265 bp (deletion of cadB allele) fragments amplified from TX01ΔcadB and TX01ΔcadBA genomic DNA with primers CadBKOF3/CadBKOR3, respectively. Lane 2 was 901-bp cadB fragment. Lanes 3, 6 and 9 were 16S rRNA gene fragments amplified from three mutants with universal primers 27F/1492R. M is DS 2000 marker.

Additional file 3.

The swimming ability of complementary strains TX01ΔcadAC and TX01ΔcadBC in the acid swimming plate. TX01, TX01ΔcadAC, and TX01ΔcadBC were cultured in LB medium to an OD600 of 0.5, then aliquots of cell suspensions (1 μL) were inoculated into the center of swimming plates including 0.3% (W/V) agar with pH = 5.5 at 30 °C for 18 h (A). The diameter of the swimming zone from the swimming plate at pH = 5.5 (B). The data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01.

Additional file 4.

The expression of acid resistance genes in Edwardsiella tarda under acidic conditions. The exponential phase of overnight cultures of TX01 were grown in normal LB medium at pH 7.0 and acidic medium at pH 5.5 for 1 h. The relative expression of ETAE_3006 and ETAE_1884 were determined by RT-qPCR. The fold difference derived from the values under acidic conditions compared with the values under normal conditions. Data are presented as the means ± SEM (N = 3). N, the number of times the experiment was performed. **, P < 0.01.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, C., Huo, X., Gu, H. et al. Acid resistance system CadBA is implicated in acid tolerance and biofilm formation and is identified as a new virulence factor of Edwardsiella tarda. Vet Res 52, 117 (2021). https://doi.org/10.1186/s13567-021-00987-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13567-021-00987-x

Keywords