Researching bacterial pathogenesis

 

 

 

 

 

 

S-Nitrosylation of the virulence regulator AphB promotes Vibrio cholerae pathogenesis

Chen, et al.

PLoS Pathogens 2022

Abstract

Vibrio cholerae is the etiologic agent of the severe human diarrheal disease cholera. To colonize mammalian hosts, this pathogen must defend against host-derived toxic compounds, such as nitric oxide (NO) and NO-derived reactive nitrogen species (RNS). RNS can covalently add an NO group to a reactive cysteine thiol on target proteins, a process called protein S-nitrosylation, which may affect bacterial stress responses. To better understand how V. cholerae regulates nitrosative stress responses, we profiled V. cholerae protein S-nitrosylation during RNS exposure. We identified an S-nitrosylation of cysteine 235 of AphB, a LysR-family transcription regulator that activates the expression of tcpP, which activates downstream virulence genes. Previous studies show that AphB C235 is sensitive to O2 and reactive oxygen species (ROS). Under microaerobic conditions, AphB formed dimer and directly repressed transcription of hmpA, encoding a flavohemoglobin that is important for NO resistance of V. cholerae. We found that tight regulation of hmpA by AphB under low nitrosative stress was important for V. cholerae optimal growth. In the presence of NO, S-nitrosylation of AphB abolished AphB activity, therefore relieved hmpA expression. Indeed, non-modifiable aphBC235S mutants were sensitive to RNS in vitro and drastically reduced colonization of the RNS-rich mouse small intestine. Finally, AphB S-nitrosylation also decreased virulence gene expression via debilitation of tcpP activation, and this regulation was also important for V. cholerae RNS resistance in vitro and in the gut. These results suggest that the modulation of the activity of virulence gene activator AphB via NO-dependent protein S-nitrosylation is critical for V. cholerae RNS resistance and colonization..

 

 

A commensal-encoded genotoxin drives restriction of Vibrio cholerae colonization and host gut microbiome remodeling

Chen, et al.

PNAS 2022

Abstract

Members of complex microbial communities that reside in environments such as the mammalian gut have evolved mechanisms of interspecies competition, which may be directed at resident microbial and host cells. While previous work has focused mainly on metabolic or niche competition, few specific intermicrobial targeting mechanisms have been elucidated in the mammalian gut. Here, we show that a genotoxin produced by commensal Escherichia coli, colibactin, which was previously shown to induce DNA damage in host intestinal cells, is also able to target via a contact-dependent mechanism a variety of enteric pathogens and commensals, including the important human diarrheal pathogen Vibrio cholerae. We find that colibactin-mediated killing depends on accumulation of intracellular reactive oxygen species, leading to DNA damage and loss of target cell fitness. We also show that the presence of colibactin is associated with cholera outcomes in a large human metagenomic dataset and that colibactin can shape the microbiome by species-specific targeting of a common human gut-associated microbe Bacteroides fragilis, suggesting that genotoxin-mediated mechanisms may have broad effects in the complex polymicrobial interactions that shape commensal microbial communities and their effects on host health and disease.

 

Thiol-based functional mimicry of phosphorylation of the two-component system response regulator ArcA promotes pathogenesis in enteric pathogens

Zhou, et al.

Cell Reports 2021

Abstract

Pathogenic bacteria can rapidly respond to stresses such as reactive oxygen species (ROS) using reversible redox-sensitive oxidation of cysteine thiol (-SH) groups in regulators. Here, we use proteomics to profile reversible ROS-induced thiol oxidation in Vibrio cholerae, the etiologic agent of cholera, and identify two modified cysteines in ArcA, a regulator of global carbon oxidation that is phosphorylated and activated under low oxygen. ROS abolishes ArcA phosphorylation but induces the formation of an intramolecular disulfide bond that promotes ArcA-ArcA interactions and sustains activity. ArcA cysteines are oxidized in cholera patient stools, and ArcA thiol oxidation drives in vitro ROS resistance, colonization of ROS-rich guts, and environmental survival. In other pathogens, such as Salmonella enterica, oxidation of conserved cysteines of ArcA orthologs also promotes ROS resistance, suggesting a common role for ROS-induced ArcA thiol oxidation in modulating ArcA activity, allowing for a balance of expression of stress- and pathogenesis-related genetic programs.

 

Pathogenicity and virulence regulation of Vibrio cholerae at the interface of host-gut microbiome interactions

Ansel Hsiao and Jun Zhu

Virulence 2020

Abstract

The Gram-negative bacterium Vibrio cholerae is responsible for the severe diarrheal pandemic disease cholera, representing a major global public health concern. This pathogen transitions from aquatic reservoirs into epidemics in human populations, and has evolved numerous mechanisms to sense this transition in order to appropriately regulate its gene expression for infection. At the intersection of pathogen and host in the gastrointestinal tract lies the community of native gut microbes, the gut microbiome. It is increasingly clear that the diversity of species and biochemical activities within the gut microbiome represents a driver of infection outcome, through their ability to manipulate the signals used by V. cholerae to regulate virulence and fitness in vivo. A better mechanistic understanding of how commensal microbial action interacts with V. cholerae pathogenesis may lead to novel prophylactic and therapeutic interventions for cholera. Here, we review a subset of this burgeoning field of research.

 

Siderophore piracy enhances Vibrio cholerae environmental survival and pathogenesis

Hyuntae Byun, I-Ji Jung, Jiandong Chen, Jessie Larios Valencia, Jay Zhu

Microbiology 2020

Abstract

Vibrio cholerae, the aetiological agent of cholera, possesses multiple iron acquisition systems, including those for the transport of siderophores. How these systems benefit V. cholerae in low-iron, polymicrobial communities in environmental settings or during infection remains poorly understood. Here, we demonstrate that in iron-limiting conditions, co-culture of V. cholerae with a number of individual siderophore-producing microbes significantly promoted V. cholerae growth in vitro. We further show that in the host environment with low iron, V. cholerae colonizes better in adult mice in the presence of the siderophore-producing commensal Escherichia coli. Taken together, our results suggest that in aquatic reservoirs or during infection, V. cholerae may overcome environmental and host iron restriction by hijacking siderophores from other microbes.

 

Vibrio cholerae Virulence Activator ToxR Regulates Manganese Transport and Resistance to Reactive Oxygen Species

Hang-Hang Jiang, Yitian Zhou, Ming Liu, Jessie Larios-Valencia, Zachariah Lee, Hui Wang, Xing-Hua Gao, Jun Zhu

Infect Immun. 2020

Abstract

Like many other pathogens, Vibrio cholerae, the causative agent of cholera, can modulate its gene expression to combat stresses encountered in both aquatic and host environments, including stress posed by reactive oxygen species (ROS). We previously reported that the virulence activator AphB in V. cholerae is involved in ROS resistance. In this study, we found that another key virulence regulator, ToxR, was important for V. cholerae resistance to hydrogen peroxide. Through a genome-wide transposon screen, we discovered that a deletion in mneA, which encodes a manganese exporter, restored ROS resistance of the toxR mutant. We then showed that ToxR did not affect mneA transcription but that the ToxR-regulated major porin OmpU was critical for ROS resistance. The addition of manganese in culture medium restored ROS resistance in both the toxR and ompU mutants. Furthermore, elemental analysis indicated that the intracellular concentration of manganese in both the toxR and ompU mutants was reduced. This may result in intracellular ROS accumulation in these mutants. Our data suggest that ToxR plays an important role in the resistance to reactive oxygen species through the regulation of manganese transport.

 

Colistin-resistance-mediated bacterial surface modification sensitizes phage infection.

Hao G, Chen AI, Liu M, Zhou H, Egan M, Yang X, Kan B, Wang H, Goulian M, Zhu J.

Antimicrob Agents Chemother 2019

Abstract

Colistin is a drug of last resort for the treatment of many multidrug resistant Gram-negative bacteria, including Klebsiella pneumoniae. However, bacteria readily acquire resistance to this antibiotic via lipopolysaccharide modifications caused by spontaneous mutations or from enzymes acquired by lateral gene transfer. The fitness cost associated with these modifications remains poorly understood. In this study, we show that colistin-resistant K. pneumoniae are more susceptible to killing by a newly isolated lytic phage than the colistin sensitive parent strain. We observe this behavior for colistin-resistance conferred by a horizontally transferred mcr-1 containing plasmid and also from the inactivation of the chromosomal gene mgrB. By measuring zeta potentials, we found that the phage particles were negatively charged at neutral pH and that colistin-resistant bacteria had less negative zeta potentials than did wildtype. These results suggest that the decreased negative surface charge of colistin-resistant cells lowers the electrostatic repulsion between the phage and bacteria, thereby promoting phage adherence and subsequent infection. To further explore this, we tested the effect of phage treatment on K. pneumoniae growing in several different environments. We found that colistin-resistant cells were more susceptible to phage than were the wildtype cells when growing in biofilms or infected moth larvae and when colonizing the mammalian gut. A better understanding of these fitness costs may lead to new treatment approaches that minimize the emergence and spread of colistin-resistant pathogens in human and environmental reservoirs.

 

CitAB Two-Component System-Regulated Citrate Utilization Contributes to Vibrio cholerae Competitiveness with the Gut Microbiota.

Liu M, Hao G, Li Z, Zhou Y, Garcia-Sillas R, Li J, Wang H, Kan B, Zhu J.

Infect Immunity 2019

Abstract

Citrate is a ubiquitous compound and can be utilized by many bacterial species, including enteric pathogens, as a carbon and energy source. Genes involved in citrate utilization have been extensively studied in some enteric bacteria, such as Klebsiella pneumoniae; however, their role in pathogenesis is still not clear. In this study, we investigated citrate utilization and regulation in Vibrio cholerae, the causative agent of cholera. The putative anaerobic citrate fermentation genes in V. cholerae, consisting of citCDEFXG, citS-oadGAB, and the two-component system (TCS) genes citAB, are highly homologous to those in K. pneumoniae Deletion analysis shows that these cit genes are essential for V. cholerae growth when citrate is the sole carbon source. The expression of citC and citS operons was dependent on citrate and CitAB, whose transcription was autorepressed and regulated by another TCS regulator, ArcA. In addition, citrate fermentation was under the control of catabolite repression. Mouse colonization experiments showed that V. cholerae can utilize citrate in vivo using the citrate fermentation pathway and that V. cholerae likely needs to compete with other members of the gut microbiota to access citrate in the gut.

 

Hypermutation-induced in vivo oxidative stress resistance enhances Vibrio cholerae host adaptation.

Wang H, X. Xing, J. Wang, B. Pang, M. Liu, J. Larios-Valencia, T. Liu, G. Liu, S. Xie, G. Hao, Liu Z, B. Kan, Zhu J.

PLoS Pathogens 2018

Abstract

Bacterial pathogens are highly adaptable organisms, a quality that enables them to overcome changing hostile environments. For example, Vibrio cholerae, the causative agent of cholera, is able to colonize host small intestines and combat host-produced reactive oxygen species (ROS) during infection. To dissect the molecular mechanisms utilized by V. cholerae to overcome ROS in vivo, we performed a whole-genome transposon sequencing analysis (Tn-seq) by comparing gene requirements for colonization using adult mice with and without the treatment of the antioxidant, N-acetyl cysteine. We found that mutants of the methyl-directed mismatch repair (MMR) system, such as MutS, displayed significant colonization advantages in untreated, ROS-rich mice, but not in NAC-treated mice. Further analyses suggest that the accumulation of both catalase-overproducing mutants and rugose colony variants in NAC- mice was the leading cause of mutS mutant enrichment caused by oxidative stress during infection. We also found that rugose variants could revert back to smooth colonies upon aerobic, in vitro culture. Additionally, the mutation rate of wildtype colonized in NAC- mice was significantly higher than that in NAC+ mice. Taken together, these findings support a paradigm in which V. cholerae employs a temporal adaptive strategy to battle ROS during infection, resulting in enriched phenotypes. Moreover, ΔmutS passage and complementation can be used to model hypermuation in diverse pathogens to identify novel stress resistance mechanisms.

OxyR2 Modulates OxyR1 Activity and Vibrio cholerae Oxidative Stress Response.

Wang H, Naseer N, Chen Y, Zhu AY, Kuai X, Galagedera N, Liu Z, Zhu J.

Infect Immunity 2017

Abstract

Bacteria have developed capacities to deal with different stresses and adapt to different environmental niches. The human pathogen Vibrio cholerae, the causative agent of the severe diarrheal disease cholera, utilizes the transcriptional regulator OxyR to activate genes related to oxidative stress resistance, including peroxiredoxin PrxA, in response to hydrogen peroxide. In this study, we identified another OxyR homolog in V. cholerae, which we named OxyR2, and we renamed the previous OxyR OxyR1. We found that OxyR2 is required to activate its divergently transcribed gene ahpC, encoding an alkylhydroperoxide reductase, independently of H2O2 A conserved cysteine residue in OxyR2 is critical for this function. Mutation of either oxyR2 or ahpC rendered V. cholerae more resistant to H2O2 RNA sequencing analyses indicated that OxyR1-activated oxidative stress-resistant genes were highly expressed in oxyR2 mutants even in the absence of H2O2 Further genetic analyses suggest that OxyR2-activated AhpC modulates OxyR1 activity by maintaining low intracellular concentrations of H2O2 Furthermore, we showed that ΔoxyR2 and ΔahpC mutants were less fit when anaerobically grown bacteria were exposed to low levels of H2O2 or incubated in seawater. These results suggest that OxyR2 and AhpC play important roles in the V. cholerae oxidative stress response.

OxyR-activated expression of Dps is important for Vibrio cholerae oxidative stress resistance and pathogenesis.

Xia X, Larios-Valencia J, Liu Z, Xiang F, Kan B, Wang H, Zhu J.

PLoS One. 2017

Abstract

Vibrio cholerae is the causative agent of cholera, a dehydrating diarrheal disease. This Gram-negative pathogen is able to modulate its gene expression in order to combat stresses encountered in both aquatic and host environments, including stress posed by reactive oxygen species (ROS). In order to further the understanding of V. cholerae's transcriptional response to ROS, we performed an RNA sequencing analysis to determine the transcriptional profile of V. cholerae when exposed to hydrogen hydroperoxide. Of 135 differentially expressed genes, VC0139 was amongst the genes with the largest induction. VC0139 encodes a protein homologous to the DPS (DNA-binding protein from starved cells) protein family, which are widely conserved and are implicated in ROS resistance in other bacteria. Using a promoter reporter assay, we show that during exponential growth, dps is induced by H2O2 in a manner dependent on the ROS-sensing transcriptional regulator, OxyR. Upon entry into stationary phase, the major stationary phase regulator RpoS is required to transcribe dps. Deletion of dps impaired V. cholerae resistance to both inorganic and organic hydroperoxides. Furthermore, we show that Dps is involved in resistance to multiple environmental stresses. Finally, we found that Dps is important for V. cholerae adult mouse colonization, but becomes dispensable in the presence of antioxidants. Taken together, our results suggest that Dps plays vital roles in both V. cholerae stress resistance and pathogenesis.

 

Thiol-based switch mechanism of virulence regulator AphB modulates oxidative stress response in Vibrio cholerae.

Liu Z, Wang H, Zhou Z, Sheng Y, Naseer N, Kan B, Zhu J.

Mol Microbiol. 2016

Abstract

Bacterial pathogens display versatile gene expression to adapt to changing surroundings. For example, Vibrio cholerae, the causative agent of cholera, utilizes distinct genetic programs to combat reactive oxygen species (ROS) in aquatic environments or during host infection. We previously reported that the virulence activator AphB in V. cholerae is involved in ROS resistance. Here by performing a genetic screen, we show that AphB represses ROS resistance gene ohrA, which is also repressed by another regulator, OhrR. Reduced forms of both AphB and OhrR directly bind to the ohrA promoter and repress its expression, whereas organic hydroperoxides such as cumene hydroperoxide (CHP) deactivate AphB and OhrR. OhrA is critical for V. cholerae adult mouse colonization but is dispensable when the mice are treated with antioxidants. Furthermore, similar to our previous finding that AphB and OhrR exhibit different reduction rates during the shift from oxic to anoxic environments, we found that AphB is also oxidized more slowly than OhrR under peroxide stress or exposure to oxygen. This differential regulation optimizes the expression of ohrA and contributes to V. cholerae's ability to survive in a variety of environmental niches that contain different levels of ROS.

 

Calcium Enhances Bile Salt-Dependent Virulence Activation in Vibrio cholerae.

Hay AJ, Yang M, Xia X, Liu Z, Hammons J, Fenical W, Zhu J.

Infect Immun. 2016

Abstract

Vibrio cholerae is the causative bacteria of the diarrheal disease cholera, but it also persists in aquatic environments, where it displays an expression profile that is distinct from that during infection. Upon entry into the host, a tightly regulated circuit coordinates the induction of two major virulence factors: cholera toxin and a toxin-coregulated pilus (TCP). It has been shown that a set of bile salts, including taurocholate, serve as host signals to activate V. cholerae virulence through inducing the activity of the transmembrane virulence regulator TcpP. In this study, we investigated the role of calcium, an abundant mental ion in the gut, in the regulation of virulence. We show that whereas Ca2+ alone does not affect virulence, Ca2+ enhances bile salt-dependent virulence activation for V. cholerae The induction of TCP by murine intestinal contents is counteracted when Ca2+ is depleted by the high-affinity calcium chelator EGTA, suggesting that the calcium present in the gut is a relevant signal for V. cholerae virulence induction in vivo We further show that Ca2+ enhances virulence by promoting bile salt-induced TcpP-TcpP interaction. Moreover, fluorescence recovery after photobleaching (FRAP) analysis demonstrated that exposure to bile salts and Ca2+ together decreases the recovery rate for fluorescently labeled TcpP, but not for another inner membrane protein (TatA). Together, these data support a model in which physiological levels of Ca2+ may result in altered bile salt-induced TcpP protein movement and activity, ultimately leading to an increased expression of virulence.

 

Plant nodulation inducers enhance horizontal gene transfer of Azorhizobium caulinodans symbiosis island.

Ling J, Wang H, Wu P, Li T, Tang Y, Naseer N, Zheng H, Masson-Boivin C, Zhong Z, Zhu J.

Proc Natl Acad Sci U S A. 2016

Abstract

Horizontal gene transfer (HGT) of genomic islands is a driving force of bacterial evolution. Many pathogens and symbionts use this mechanism to spread mobile genetic elements that carry genes important for interaction with their eukaryotic hosts. However, the role of the host in this process remains unclear. Here, we show that plant compounds inducing the nodulation process in the rhizobium-legume mutualistic symbiosis also enhance the transfer of symbiosis islands. We demonstrate that the symbiosis island of the Sesbania rostrata symbiont, Azorhizobium caulinodans, is an 87.6-kb integrative and conjugative element (ICEAc) that is able to excise, form a circular DNA, and conjugatively transfer to a specific site of gly-tRNA gene of other rhizobial genera, expanding their host range. The HGT frequency was significantly increased in the rhizosphere. An ICEAc-located LysR-family transcriptional regulatory protein AhaR triggered the HGT process in response to plant flavonoids that induce the expression of nodulation genes through another LysR-type protein, NodD. Our study suggests that rhizobia may sense rhizosphere environments and transfer their symbiosis gene contents to other genera of rhizobia, thereby broadening rhizobial host-range specificity.

 

Differential thiol-based switches jump-start Vibrio cholerae pathogenesis.

Liu, Z., H Wang, Z Zhou, N Naseer, F Xiang, B Kan, M Goulian, and J. Zhu.

Cell Reports 2016.

Abstract

Bacterial pathogens utilize gene expression versatility to adapt to environmental changes. Vibrio cholerae, the causative agent of cholera, encounters redox-potential changes when it transitions from oxygen-rich aquatic reservoirs to the oxygen-limiting human gastrointestinal tract. We previously showed that the virulence regulator AphB uses thiol-based switches to sense the anoxic host environment and transcriptionally activate the key virulence activator tcpP. Here, by performing a high-throughput transposon sequencing screen in vivo, we identified OhrR as another regulator that enables V. cholerae rapid anoxic adaptation. Like AphB, reduced OhrR binds to and regulates the tcpP promoter. OhrR and AphB displayed differential dynamics in response to redox-potential changes: OhrR is reduced more rapidly than AphB. Furthermore, OhrR thiol modification is required for rapid activation of virulence and successful colonization. This reveals a mechanism whereby bacterial pathogens employ posttranslational modifications of multiple transcription factors to sense and adapt to dynamic environmental changes.

 

Dual Zinc uptake systems promote Vibrio cholerae competitiveness against gut microbiome.

Sheng, Y., F. Fan, O. Jessen, Z. Zhong, B. Kan, H. Wang, and J. Zhu.

Infec Immun. 2015.

Abstract

Zinc is an essential trace metal required for numerous cellular processes in all forms of life. In order to maintain zinc homeostasis, bacteria have developed several transport systems to regulate its uptake. In this study, we investigated zinc transport systems in the enteric pathogen Vibrio cholerae, the causative agent of cholera. Bioinformatic analysis predicts that two gene clusters, VC2081 to VC2083 (annotated as zinc utilization genes znuABC) and VC2551 to VC2555 (annotated as zinc-regulated genes zrgABCDE), are regulated by the putative zinc uptake regulator Zur. Using promoter reporter and biochemical assays, we confirmed that Zur represses znuABC and zrgABCDE promoters in a Zn2+-dependent manner. Under Zn2+-limiting conditions, we found that mutations in either the znuABC or zrgABCDE gene cluster affect bacterial growth, with znuABC mutants displaying a more severe growth defect, suggesting that both ZnuABC and ZrgABCDE are involved in Zn2+ uptake and that ZnuABC plays the predominant role. Furthermore, we reveal that ZnuABC and ZrgABCDE are important for V. cholerae colonization in both infant and adult mouse models, particularly in the presence of other intestinal microbiota. Collectively, our studies indicate that these two zinc transporter systems play vital roles in maintaining zinc homeostasis during V. cholerae growth and pathogenesis.

 

"Quorum Non-Sensing": Social Cheating and Deception in Vibrio cholerae.

Katzianer, D.S., H Wang, R Carey, and J. Zhu.

Appl. Environ. Microbiol. 2015.

Abstract

Quorum sensing (QS) is widely used by bacteria to coordinate behavior in response to external stimuli. In Vibrio cholerae, this process is important for environmental survival and pathogenesis, though, intriguingly, a large percentage of natural isolates are QS-deficient. Here, we show that QS-deficient mutants can spread as social cheaters by ceasing production of extracellular proteases under conditions requiring their growth. We further show that mutants stimulate biofilm formation and are over-represented in biofilms compared to planktonic communities; on this basis, we suggest that QS-deficient mutants may have the side-effect of enhancing environmental tolerance of natural populations due to the inherent resistance properties of biofilms. Interestingly, high frequencies of QS-deficient individuals did not impact production of QS signaling molecules despite mutants being unable to respond to these inducers, indicating that these variants actively cheat by false-signaling in conditions requiring QS. Taken together, our results suggest that social cheating may drive QS deficiency emergence within V. cholerae natural populations.

 

Vibrio cholerae represses polysaccharide synthesis to promote motility in mucosa.

Liu, Z., Y. Wang, S. Liu, Y Sheng, K. Rueggebert, H. Wang, J. Li, F.X. Gu, Z. Zhong, B. Kan, and J. Zhu.

Infec. Immun. 2015.

Abstract

The viscoelastic mucus layer of gastrointestinal tracts is a host defense barrier that a successful enteric pathogen, such as Vibrio cholerae, must circumvent. V. cholerae, the causative agent of cholera, is able to penetrate the mucosa and colonize the epithelial surface of the small intestine. In this study, we found that mucin, the major component of mucus, promoted V. cholerae movement on semisolid medium and in liquid medium. A genome-wide screen revealed that Vibrio polysaccharide (VPS) production was inversely correlated with mucin-enhanced motility. Mucin adhesion assays indicated that VPS bound to mucin. Moreover, we found that vps expression was reduced upon exposure to mucin. In an infant mouse colonization model, mutants that overexpressed VPS colonized less effectively than wild-type strains in more distal intestinal regions. These results suggest that V. cholerae is able to sense mucosal signals and modulate vps expression accordingly so as to promote fast motion in mucus, thus allowing for rapid spread throughout the intestines.

 

Intestinal signals-promoted biofilm dispersal stimulates Vibrio cholerae virulence gene expression.

Hay, A., and J. Zhu.

Infec. Immun. 2015.

Abstract

Vibrio cholerae causes human infection through ingestion of contaminated food and water, leading to the devastating diarrheal disease cholera. V. cholerae forms matrix-encased aggregates, known as biofilms, in the native aquatic environment. While the formation of V. cholerae biofilms has been well studied, little is known about the dispersal from biofilms, particularly upon entry into the host. In this study, we found that the exposure of mature biofilms to physiologic levels of the bile salt taurocholate, a host signal for the virulence gene induction of V. cholerae, induces an increase in the number of detached cells with a concomitant decrease in biofilm mass. Scanning electron microscopy micrographs of biofilms exposed to taurocholate revealed an altered, perhaps degraded, appearance of the biofilm matrix. The inhibition of protein synthesis did not alter rates of detachment, suggesting that V. cholerae undergoes a passive dispersal. Cell-free media from taurocholate-exposed biofilms contains a larger amount of free polysaccharide, suggesting an abiotic degradation of biofilm matrix by taurocholate. Furthermore, we found that V. cholerae is only able to induce virulence in response to taurocholate after exit from the biofilm. Thus, we propose a model in which V. cholerae ingested as a biofilm has coopted the host-derived bile salt signal to detach from the biofilm and go on to activate virulence.

 

A high-throughput small molecule screen to identify a novel chemical inhibitor of Clostridium difficile.

Katzianer, D., T. Yano, H. Rubin, and J. Zhu.

Int. J. Antimicrob. Agents. 2014.

Abstract

Clostridium difficile, a highly drug-resistant Gram-positive, spore-forming bacterium, remains a leading cause of hospital-acquired diarrhoea and antibiotic-associated colitis. Clinically, only a handful of antibiotics are used for treating C. difficile infection (CDI), suggesting a necessity for the development of new treatment options. Here we performed a high-throughput screen of 2000 drug-like compounds for inhibition of C. difficile. From this screen, one compound, 5-nitro-1,10-phenanthroline (5-NP), showed potent bactericidal effects in vitro. In addition, this compound displayed high potency towards other Clostridium spp. as well as Mycobacterium bovis but not towards other tested Gram-positive and Gram-negative bacteria. Furthermore, we show that this inhibition may proceed through a metal chelation-dependent mechanism. More importantly, preliminary evidence suggests moderate efficacy for this compound in treating CDI in a murine infection model. These results present a possible basis for the further development of this compound as an antibiotic treatment for CDI.

 

Escherichia coli isolate for studying colonization of the mouse intestine and its application to two-component signaling knockouts.

Lasaro, M., Z . Liu, R. Bishar, K. Kelly, S. Chattopadhyay, E. Sokurenko, J. Zhu*, and M. Goulian*.

J. Bacteriol. 2014.

Abstract

The biology of Escherichia coli in its primary niche, the animal intestinal tract, is remarkably unexplored. Studies with the streptomycin-treated mouse model have produced important insights into the metabolic requirements for Escherichia coli to colonize mice. However, we still know relatively little about the physiology of this bacterium growing in the complex environment of an intestine that is permissive for the growth of competing flora. We have developed a system for studying colonization using an E. coli strain, MP1, isolated from a mouse. MP1 is genetically tractable and does not require continuous antibiotic treatment for stable colonization. As an application of this system, we have separately knocked out each two-component system response regulator in MP1 and performed competitions against the wild-type parent. We find that only three response regulators, ArcA, CpxR, and RcsB, produce strong colonization defects, suggesting that in addition to anaerobiosis, adaptation to cell envelope stress is a critical requirement for E. coli colonization of the mouse intestine. We also show that the response regulator OmpR, which had previously been hypothesized to be important for adaptation between in vivo and ex vivo environments, is not required for MP1 colonization due to the presence of a third major porin.

 

Enhanced Interaction of Vibrio cholerae Virulence Regulators TcpP and ToxR under Oxygen-Limiting Conditions

Fan F., Z. Liu, N. Jabeen, L.D. Birdwell, J. Zhu and Kan B.

Infect Immun. 2014 Feb. 3.

Abstract

Vibrio cholerae is the causative agent of the diarrheal disease cholera. The ability of V. cholerae to colonize and cause disease requires the intricately regulated expression of a number of virulence factors during infection. One of the signals sensed by V. cholerae is oxygen-limiting conditions in the gut. It has been shown that the virulence activator AphB plays a key role to sense low oxygen concentration and induce the transcription of another key virulence activator, TcpP. In this study, we applied a bacterial two-hybrid system to further examine the oxygen effect on different virulence regulators. We found that anoxic conditions enhanced interaction between TcpP and ToxR, identified as the first positive regulator of V. cholerae virulence genes. We further demonstrated that the TcpP-ToxR interaction was dependent on the primary periplasmic protein disulfide formation enzyme DsbA and cysteine residues in the periplasmic domains of both ToxR and TcpP. Furthermore, we showed that in V. cholerae, an interaction between TcpP-ToxR is important for virulence gene induction. Under anaerobic growth conditions, we detected ToxR-TcpP heterodimers, which were abolished in the presence of the reducing agent DTT. Our results suggest that V. cholerae may sense intestinal anoxic signals by multiple components to activate virulence.

 

 

A novel protein protects bacterial iron-dependent metabolism from nitric oxide.

Stern, A.M., B. Liu, L.R. Bakken, J.P. Shapleigh, and Zhu J.

J. Bacteriol. 195:4702-4708, 2013

Abstract

Reactive nitrogen species (RNS), in particular nitric oxide (NO), are toxic to bacteria, and bacteria have mechanisms to allow growth despite this stress. Understanding how bacteria interact with NO is essential to understanding bacterial physiology in many habitats, including pathogenesis; however, many targets of NO and enzymes involved in NO resistance remain uncharacterized. We performed for the first time a metabolomic screen on NO-treated and -untreated bacteria to define broadly the effects of NO on bacterial physiology, as well as to identify the function of NnrS, a previously uncharacterized enzyme involved in defense against NO. We found many known and novel targets of NO. We also found that iron-sulfur cluster enzymes were preferentially inhibited in a strain lacking NnrS due to the formation of iron-NO complexes. We then demonstrated that NnrS is particularly important for resistance to nitrosative stress under anaerobic conditions. Our data thus reveal the breadth of the toxic effects of NO on metabolism and identify the function of an important enzyme in alleviating this stress.

 

Quorum sensing regulatory cascades control Vibrio fluvialis pathogenesis.

Wang, Y., H. Wang, W. Liang, A. J. Hay, Z. Zhong, B. Kan, and Zhu J.

J. Bacteriol. 195:3583-3589, 2013

Abstract

Quorum sensing (QS) is a process by which individual bacteria are able to communicate with one another, thereby enabling the population as a whole to coordinate gene regulation and subsequent phenotypic outcomes. Communication is accomplished through production and detection of small molecules in the extracellular milieu. In many bacteria, particularly Vibrio species, multiple QS systems result in multiple signals, as well as cross talk between systems. In this study, we identify two QS systems in the halophilic enteric pathogen Vibrio fluvialis: one acyl-homoserine lactone (AHL) based and one CAI-1/AI-2 based. We show that a LuxI homolog, VfqI, primarily produces 3-oxo-C10-HSL, which is sensed by a LuxR homolog, VfqR. VfqR-AHL is required to activate vfqI expression and autorepress vfqR expression. In addition, we have shown that similar to that in V. cholerae and V. harveyi, V. fluvialis produces CAI-1 and AI-2 signal molecules to activate the expression of a V. cholerae HapR homolog through LuxO. Although VfqR-AHL does not regulate hapR expression, HapR can repress vfqR transcription. Furthermore, we found that QS in V. fluvialis positively regulates production of two potential virulence factors, an extracellular protease and hemolysin. QS also affects cytotoxic activity against epithelial tissue cultures. These data suggest that V. fluvialis integrates QS regulatory pathways to play important physiological roles in pathogenesis.

 

Applying reversible mutations of nodulation and nitrogen-fixation genes to study social cheating in Rhizobium etli-legume interaction.

Ling, J., H. Zheng, D.S. Katzianer, H. Wang, Z. Zhong, and Zhu, J.

PLoS One.8:e70138, 2013

Abstract

Mutualisms are common in nature, though these symbioses can be quite permeable to cheaters in situations where one individual parasitizes the other by discontinuing cooperation yet still exploits the benefits of the partnership. In the Rhizobium-legume system, there are two separate contexts, namely nodulation and nitrogen fixation processes, by which resident Rhizobium individuals can benefit by cheating. Here, we constructed reversible and irreversible mutations in key nodulation and nitrogen-fixation pathways of Rhizobium etli and compared their interaction with plant hosts Phaseolus vulgaris to that of wild type. We show that R. etli reversible mutants deficient in nodulation factor production are capable of intra-specific cheating, wherein mutants exploit other Rhizobium individuals capable of producing these factors. Similarly, we show that R. etli mutants are also capable of cheating inter-specifically, colonizing the host legume yet contributing nothing to the partnership in terms of nitrogen fixation. Our findings indicate that cheating is possible in both of these frameworks, seemingly without damaging the stability of the mutualism itself. These results may potentially help explain observations suggesting that legume plants are commonly infected by multiple bacterial lineages during the nodulation process.

 

Bile-salt induced intermolecular disulfide bond formation activates Vibrio cholerae virulence.

Yang, M., Z. Liu, C. Hughes, A.M. Stern, H. Wang, Z. Zhong, B. Kan, W. Fenical and Zhu, J.

Proc. Natl. Acad. Sci U.S.A. 110:2348-2353, 2013.

Abstract

To be successful pathogens, bacteria must often restrict the expression of virulence genes to host environments. This requires a physical or chemical marker of the host environment as well as a cognate bacterial system for sensing the presence of a host to appropriately time the activation of virulence. However, there have been remarkably few such signal-sensor pairs identified, and the molecular mechanisms for host-sensing are virtually unknown. By directly applying a reporter strain of Vibrio cholerae, the causative agent of cholera, to a thin layer chromatography (TLC) plate containing mouse intestinal extracts, we found two host signals that activate virulence gene transcription. One of these was revealed to be the bile salt taurocholate. We then show that a set of bile salts cause dimerization of the transmembrane transcription factor TcpP by inducing intermolecular disulfide bonds between cysteine (C)-207 residues in its periplasmic domain. Various genetic and biochemical analyses led us to propose a model in which the other cysteine in the periplasmic domain, C218, forms an inhibitory intramolecular disulfide bond with C207 that must be isomerized to form the active C207-C207 intermolecular bond. We then found bile salt-dependent effects of these cysteine mutations on survival in vivo, correlating to our in vitro model. Our results are a demonstration of a mechanism for direct activation of the V. cholerae virulence cascade by a host signal molecule. They further provide a paradigm for recognition of the host environment in pathogenic bacteria through periplasmic cysteine oxidation.

 

LysR-family activator regulated major facilitator superfamily transporters are involved in Vibrio cholerae antimicrobial compound resistance and intestinal colonization.

Chen, S., H. Wang, D.S. Katzianer, Z. Zhong, and Zhu, J.

Int. J. Antimicrob. Agents 41:188-192, 2013.

Abstract

Expression of a multidrug resistance transporter renders bacterial cells resistant to a variety of drugs. The major facilitator superfamily (MFS) comprises the largest group of bacterial multidrug transporters. There are over 20 MFS efflux pumps annotated on the genome of Vibrio cholerae, but little is known about their functions and regulation. In this study, five MFS efflux pumps were characterised, each of which is associated with a divergently transcribed putative LysR-type transcriptional regulator (MfsR). It was found that each of these MFS structural genes is regulated by the corresponding MfsR regulator. Deletion of these five mfs genes results in increased susceptibility to tetracycline and crude bile as well as a colonisation defect in an infant mouse colonisation model. Moreover, tetracycline and unknown intestinal signals could serve as co-inducers for the MfsR regulators. These data suggest that MFS efflux pumps are important both for antimicrobial resistance and V. cholerae pathogenesis.

 

Catalases promote resistance of oxidative stress in Vibrio cholerae.

Wang, H., S. Chen, J. Zhang, F.P. Rothenbacher, T. Jiang, B. Kan, Z. Zhong, and Zhu, J.

PLoS One.7:e53383, 2012

Abstract

Oxidative stress is a major challenge faced by bacteria. Many bacteria control oxidative stress resistance pathways through the transcriptional regulator OxyR. The human pathogen Vibrio cholerae is a Gram-negative bacterium that is the causative agent of cholera. V. cholerae lives in both aquatic environments and human small intestines, two environments in which it encounters reactive oxygen species (ROS). To study how V. cholerae responds to oxidative stress, we constructed an in-frame oxyR deletion mutant. We found that this mutant was not only sensitive to H(2)O(2), but also displayed a growth defect when diluted in rich medium. Further study showed that two catalases, KatG and KatB, either when expressed in living cells, present in culture supernatants, or added as purified recombinant proteins, could rescue the oxyR growth defect. Furthermore, although it could colonize infant mouse intestines similar to that of wildtype, the oxyR mutant was defective in zebrafish intestinal colonization. Alternatively, co-infection with wildtype, but not katG-katB deletion mutants, greatly enhanced oxyR mutant colonization. Our study suggests that OxyR in V. cholerae is critical for antioxidant defense and that the organism is capable of scavenging environmental ROS to facilitate population growth.

 

Succession in the Gut Microbiome following Antibiotic and Antibody Therapies for Clostridium difficile.

Peterfreund, G.L., L.E. Vandivier, R. Sinha, A.J. Marozsan, W.C. Olson, J. Zhu, and Bushman F.D.

PLoS One.7:e46966, 2012

Abstract

Antibiotic disruption of the intestinal microbiota may cause susceptibility to pathogens that is resolved by progressive bacterial outgrowth and colonization. Succession is central to ecological theory but not widely documented in studies of the vertebrate microbiome. Here, we study succession in the hamster gut after treatment with antibiotics and exposure to Clostridium difficile. C. difficile infection is typically lethal in hamsters, but protection can be conferred with neutralizing antibodies against the A and B toxins. We compare treatment with neutralizing monoclonal antibodies (mAb) to treatment with vancomycin, which prolongs the lives of animals but ultimately fails to protect them from death. We carried out longitudinal deep sequencing analysis and found distinctive waves of succession associated with each form of treatment. Clindamycin sensitization prior to infection was associated with the temporary suppression of the previously dominant Bacteroidales and the fungus Saccinobaculus in favor of Proteobacteria. In mAb-treated animals, C. difficile proliferated before joining Proteobacteria in giving way to re-expanding Bacteroidales and the fungus Wickerhamomyces. However, the Bacteroidales lineages returning by day 7 were different from those that were present initially, and they persisted for the duration of the experiment. Animals treated with vancomycin showed a different set of late-stage lineages that were dominated by Proteobacteria as well as increased disparity between the tissue-associated and luminal cecal communities. The control animals showed no change in their gut microbiota. These data thus suggest different patterns of ecological succession following antibiotic treatment and C. difficile infection.

 

The NorR regulon is critical for Vibrio cholerae resistance to nitric oxide and sustained colonization of the intestines.

Stern AM, Hay AJ, Liu Z, Desland FA, Zhang J, Zhong Z, Zhu J.

MBio. 2012 Apr 17;3(2):e00013-12. doi: 10.1128/mBio.00013-12. Print 2012.

Abstract

Vibrio cholerae, the cause of an often fatal infectious diarrhea, remains a large global public health threat. Little is known about the challenges V. cholerae encounters during colonization of the intestines, which genes are important for overcoming these challenges, and how these genes are regulated. In this study, we examined the V. cholerae response to nitric oxide (NO), an antibacterial molecule derived during infection from various sources, including host inducible NO synthase (iNOS). We demonstrate that the regulatory protein NorR regulates the expression of NO detoxification genes hmpA and nnrS, and that all three are critical for resisting low levels of NO stress under microaerobic conditions in vitro. We also show that prxA, a gene previously thought to be important for NO detoxification, plays no role in NO resistance under microaerobic conditions and is upregulated by H(2)O(2), not NO. Furthermore, in an adult mouse model of prolonged colonization, hmpA and norR were important for the resistance of both iNOS- and non-iNOS-derived stresses. Our data demonstrate that NO detoxification systems play a critical role in the survival of V. cholerae under microaerobic conditions resembling those of an infectious setting and during colonization of the intestines over time periods similar to that of an actual V. cholerae infection. IMPORTANCE: Little is known about what environmental stresses Vibrio cholerae, the etiologic agent of cholera, encounters during infection, and even less is known about how V. cholerae senses and counters these stresses. Most prior studies of V. cholerae infection relied on the 24-h infant mouse model, which does not allow the analysis of survival over time periods comparable to that of an actual V. cholerae infection. In this study, we used a sustained mouse colonization model to identify nitric oxide resistance as a function critical for the survival of V. cholerae in the intestines and further identified the genes responsible for sensing and detoxifying this stress.

 

The VarS/VarA two-component system modulates the activity of the Vibrio cholerae quorum-sensing transcriptional regulator HapR.

Tsou AM, Liu Z, Cai T, Zhu J.

Microbiology. 2011 Jun; 157(Pt 6):1620-8. Epub 2011 Mar 10.

Abstract

The human pathogen Vibrio cholerae uses quorum sensing to regulate the expression of a number of phenotypes, including virulence factor production, in response to changes in cell density. It produces small molecules called autoinducers that increase in concentration as cell density increases, and these autoinducers bind to membrane sensors once they reach a certain threshold. This binding leads to signalling through a downstream phosphorelay pathway to alter the expression of the transcriptional regulator HapR. Previously, it was shown that the VarS/VarA two-component system acts on a component of the phosphorelay pathway upstream of HapR to regulate HapR expression levels. Here, we show that in addition to this mechanism of regulation, VarS and VarA also indirectly modulate HapR protein activity. This modulation is mediated by the small RNA CsrB but is independent of the known quorum-sensing system that links the autoinducers to HapR. Thus, the VarS/VarA two-component system intersects with the quorum-sensing network at two levels. In both cases, the effect of VarS and VarA on quorum sensing is dependent on the Csr small RNAs, which regulate carbon metabolism, suggesting that V. cholerae may integrate nutrient status and cell density sensory inputs to tailor its gene expression profile more precisely to surrounding conditions.

Vibrio cholerae anaerobic induction of virulence gene expression is controlled by thiol-based switches of virulence regulator AphB

Liu Z, Yang M, Peterfreund GL, Tsou AM, Selamoglu N, Daldal F, Zhong Z, Kan B, Zhu J.

Proc Natl Acad Sci U.S.A. 2011 Jan 11;108(2):810-5. Epub 2010 Dec 27.

Abstract

Bacterial pathogens have evolved sophisticated signal transduction systems to coordinately control the expression of virulence determinants. For example, the human pathogen Vibrio cholerae is able to respond to host environmental signals by activating transcriptional regulatory cascades. The host signals that stimulate V. cholerae virulence gene expression, however, are still poorly understood. Previous proteomic studies indicated that the ambient oxygen concentration plays a role in V. cholerae virulence gene expression. In this study, we found that under oxygen-limiting conditions, an environment similar to the intestines, V. cholerae virulence genes are highly expressed. We show that anaerobiosis enhances dimerization and activity of AphB, a transcriptional activator that is required for the expression of the key virulence regulator TcpP, which leads to the activation of virulence factor production. We further show that one of the three cysteine residues in AphB, C(235), is critical for oxygen responsiveness, as the AphB(C235S) mutant can activate virulence genes under aerobic conditions in vivo and can bind to tcpP promoters in the absence of reducing agents in vitro. Mass spectrometry analysis suggests that under aerobic conditions, AphB is modified at the C(235) residue. This modification is reversible between oxygen-rich aquatic environments and oxygen-limited human hosts, suggesting that V. cholerae may use a thiol-based switch mechanism to sense intestinal signals and activate virulence.

Virulence regulator AphB enhances toxR transcription in Vibrio cholerae.

Xu X, Stern AM, Liu Z, Kan B, Zhu J.

BMC Microbiol. 2010 Jan 6;10:3.

Abstract

BACKGROUND:

Vibrio cholerae is the causative agent of cholera. Extensive studies reveal that complicated regulatory cascades regulate expression of virulence genes, the products of which are required for V. cholerae to colonize and cause disease. In this study, we investigated the expression of the key virulence regulator ToxR under different conditions.

RESULTS:

We found that compared to that of wild type grown to stationary phase, the toxR expression was lower in an aphB mutant strain. AphB has been previously shown to be a key virulence regulator that is required to activate the expression of tcpP. When expressed constitutively, AphB is able to activate the toxR promoter. Furthermore, gel shift analysis indicates that AphB binds toxR promoter region directly. We also characterize the effect of AphB on the levels of the outer membrane porins OmpT and OmpU, which are known to be regulated by ToxR.

CONCLUSIONS:

Our data indicate that V. cholerae possesses an additional regulatory loop that use AphB to activate the expression of two virulence regulators, ToxR and TcpP, which together control the expression of the master virulence regulator ToxT.

Metabolism of bile salts in mice influences spore germination in Clostridium difficile.

Giel JL, Sorg JA, Sonenshein AL, Zhu J.

PLoS One. 2010 Jan 15;5(1):e8740.

Abstract

Clostridium difficile, a spore-forming bacterium, causes antibiotic-associated diarrhea. In order to produce toxins and cause disease, C. difficile spores must germinate and grow out as vegetative cells in the host. Although a few compounds capable of germinating C. difficile spores in vitro have been identified, the in vivo signal(s) to which the spores respond were not previously known. Examination of intestinal and cecal extracts from untreated and antibiotic-treated mice revealed that extracts from the antibiotic-treated mice can stimulate colony formation from spores to greater levels. Treatment of these extracts with cholestyramine, a bile salt binding resin, severely decreased the ability of the extracts to stimulate colony formation from spores. This result, along with the facts that the germination factor is small, heat-stable, and water-soluble, support the idea that bile salts stimulate germination of C. difficile spores in vivo. All extracts able to stimulate high level of colony formation from spores had a higher proportion of primary to secondary bile salts than extracts that could not. In addition, cecal flora from antibiotic-treated mice was less able to modify the germinant taurocholate relative to flora from untreated mice, indicating that the population of bile salt modifying bacteria differed between the two groups. Taken together, these data suggest that an in vivo-produced compound, likely bile salts, stimulates colony formation from C. difficile spores and that levels of this compound are influenced by the commensal gastrointestinal flora.

 

The virulence transcriptional activator AphA enhances biofilm formation by Vibrio cholerae by activating expression of the biofilm regulator VpsT.

Yang M, Frey EM, Liu Z, Bishar R, Zhu J.

Infect Immun. 2010 Feb;78(2):697-703. Epub 2009 Nov 23.

Abstract

Vibrio cholerae is the agent of the severe diarrheal disease cholera, and it perpetuates in aquatic reservoirs when not in the host. Within the host's intestines, the bacteria execute a complex regulatory pathway culminating with the production of virulence factors that allow colonization and cause disease. The ability of V. cholerae to form biofilms is thought to aid its persistence in the aquatic environment and passage through the gastric acid barrier of the stomach. The transcriptional activators VpsR and VpsT are part of the biofilm formation-regulatory network. In this study, we screened a V. cholerae genomic library in Escherichia coli cells containing a P(vpsT)-luxCDBAE transcriptional fusion reporter and found that a plasmid clone containing the aphA gene activates the expression of vpsT in E. coli. AphA is a master virulence regulator in V. cholerae that is required to activate the expression of tcpP, whose gene products in turn activate all virulence genes including those responsible for the synthesis of the toxin-coregulated pilus (TCP) and cholera toxin through the activation of toxT. AphA has a direct effect on the vpsT promoter, as gel shift experiments demonstrated that AphA binds to the vpsT promoter region. Furthermore, V. cholerae aphA mutants exhibit significantly lower levels of vpsT expression as well as reduced biofilm formation. AphA thus links the expression of virulence and biofilm synthesis genes.

Quorum sensing negatively regulates hemolysin transcriptionally and posttranslationally in Vibrio cholerae.

Tsou AM, Zhu J.

Infect Immun. 2010 Jan; 78(1):461-7. Epub 2009 Oct 26.

Abstract

Recent work has shown that in addition to cholera toxin (CT) and the toxin-coregulated pilus (TCP), other cytotoxic proteins in Vibrio cholerae also cause disease symptoms, and this is particularly evident in strains lacking CT. One such protein is the hemolysin encoded by hlyA. Here we show that, like CT and TCP, HlyA is repressed by the quorum-sensing-regulated transcription factor HapR. This repression occurs on two levels: one at the transcriptional level that is independent of the metalloprotease HapA and one at the posttranslational level that is mediated by HapA. The transcriptional regulation is significantly more apparent on solid media than in liquid cultures. This is the first time that hemolysis has been shown to be directly regulated by quorum sensing in V. cholerae, and it is interesting that, like other virulence factors, HlyA is also repressed by HapR, which is expressed late in infection.

Coordinated regulation of virulence by quorem sensing and motility pathways during the initial stages of Vibrio cholerae infection.

Tsou AM, Frey EM, Hsiao A, Liu Z, Zhu J.

Commun Integr Biol. 2008;1(1):42-4

Abstract

Pathogenic bacteria, such as Vibrio cholerae, must be capable of adapting to diverse living conditions, especially when transitioning from life in environmental reservoirs to life in a host. The abilities to sense arrival at a site suitable for colonization or infection and to respond with appropriate alterations in gene expression are crucial for a pathogen's success. Recently, we have shown that V. cholerae is able to recognize that it has reached its colonization site in the small intestine by sensing breakage of its flagellum as it penetrates the mucosal layer overlaying the intestinal epithelium. Flagellar loss results in the release of the anti-sigma factor FlgM and subsequent activation of the alternative sigma-factor FliA. FliA represses the quorum sensing-controlled transcriptional regulator, HapR, allowing increased expression of virulence factors such as Cholera Toxin (CT) and the Toxin Coregulated Pilus (TCP). In this way, the de-repression of virulence factor expression coincides with the arrival of bacteria at the site of infection at the intestinal mucosa. Our work reveals an interesting interplay between motility and quorum sensing signaling pathways to precisely time virulence gene expression during colonization.

Regulatory targets of quorem sensing in Vibrio cholerae: evidence for two distinct HapR-binding motifs.

Tsou AM, Cai T, Liu Z, Zhu J, Kulkarni RV.

Nucleic Acids Res. 2009 May;37(8):2747-56. Epub 2009 Mar 10.

Abstract

The quorum-sensing pathway in Vibrio cholerae controls the expression of the master regulator HapR, which in turn regulates several important processes such as virulence factor production and biofilm formation. While HapR is known to control several important phenotypes, there are only a few target genes known to be transcriptionally regulated by HapR. In this work, we combine bioinformatic analysis with experimental validation to discover a set of novel direct targets of HapR. Our results provide evidence for two distinct binding motifs for HapR-regulated genes in V. cholerae. The first binding motif is similar to the motifs recently discovered for orthologs of HapR in V. harveyi and V. vulnificus. However, our results demonstrate that this binding motif can be of variable length in V. cholerae. The second binding motif shares common elements with the first motif, but is of fixed length and lacks dyad symmetry at the ends. The contributions of different bases to HapR binding for this second motif were demonstrated using systematic mutagenesis experiments. The current analysis presents an approach for systematically expanding our knowledge of the quorum-sensing regulon in V. cholerae and other related bacteria.

Genetic tools to study gene expression during bacterial pathogen infection.

Hsiao A, Zhu J.

Adv Appl Microbiol. 2009;67:297-314

Abstract

The study of bacterial pathogenesis is in many ways the study of the regulatory mechanisms at work in the microbe during infection. The astonishing flexibility and adaptability of the bacterial cell has enabled many pathogenic species to freely transition between dramatically different environmental conditions. The transcriptional changes that underlie this ability can determine the success of the pathogen in the host. Many techniques have been devised to examine the transcriptional repertoire of bacteria in vivo during infection. Here, we review a class of technologies known as in vivo expression technology (IVET), which use promoter-trapping with a variety of different reporter constructs to allow researchers to probe the transcriptional changes taking place in bacteria under various environmental conditions. Using IVET techniques, researchers have been able to catalogue a wide variety of virulence factors in the host for several important human pathogens, as well as examining the timing of virulence gene regulation. Most recently, IVET techniques have also been used to identify transcriptional repression events in vivo, such as the suppression of anti-colonization factors deleterious to infection. As the array of IVET reporters and promoter-trapping strategies grow, researchers are increasingly able to illuminate the myriad transcriptional activities that allow bacteria to survive and cause disease in the host.

Direct regulation by the Vibrio cholerae regulator ToxT to modulate colonization and anticolonization pilus expression.

Hsiao A, Xu X, Kan B, Kulkarni RV, Zhu J.

Infect Immun. 2009 Apr;77(4):1383-8. Epub 2009 Jan 21.

Abstract

The pathogen Vibrio cholerae uses a large number of coordinated transcriptional regulatory events to transition from its environmental reservoir to the host and establish itself at its preferred colonization site at the host intestinal mucosa. The key regulator in this process is the AraC/XylS family transcription factor, ToxT, which plays critical roles in pathogenesis, including the regulation of two type IV pili, the anticolonization factor mannose-sensitive hemagglutinin and the toxin-coregulated pilus. Previously, it was thought ToxT required dimerization in order to effect transcriptional regulation at its cognate promoters. Here, we present evidence that ToxT directly represses transcription of the msh operon by binding to three promoters within this operon and that dimerization may not be required for transcriptional repression of target promoters by ToxT, suggesting that this regulator uses different mechanisms to modulate the transcriptional repertoire of V. cholerae.

Mucosal penetration primes Vibrio cholerae for host colonization by repressing quorum sensing

Liu Z, Miyashiro T, Tsou A, Hsiao A, Goulian M, Zhu J.

Proc Natl Acad Sci U.S.A. 2008 July 15;105(28):9769-74. Epub 2008 Jul 7.

Abstract

To successfully infect a host and cause the diarrheal disease cholera, Vibrio cholerae must penetrate the intestinal mucosal layer and express virulence genes. Previous studies have demonstrated that the transcriptional regulator HapR, which is part of the quorum sensing network in V. cholerae, represses the expression of virulence genes. Here, we show that hapR expression is also modulated by the regulatory network that governs flagellar assembly. Specifically, FliA, which is the alternative sigma-factor (sigma(28)) that activates late-class flagellin genes in V. cholerae, represses hapR expression. In addition, we show that mucin penetration by V. cholerae is sufficient to break flagella and so cause the secretion of FlgM, the anti-sigma factor that inhibits FliA activity. During initial colonization of host intestinal tissue, hapR expression is repressed because of low cell density. However, full repression of hapR expression does not occur in fliA mutants, which results in attenuated colonization. Our results suggest that V. cholerae uses flagellar machinery to sense particular intestinal signals before colonization and enhance the expression of virulence genes by modulating the output of quorum sensing signaling.

Post-transcriptional cross-talk between pro- and anti-colonization pili biosynthesis systems in Vibrio cholerae.

 

Hsiao A, Toscano K, Zhu J.

Mol Microbiol. 2008 Feb;67(4):849-60. Epub 2007 Dec 19.

Abstract

The pathogen Vibrio cholerae modulates the expression of many genes in order to transition from its environmental reservoir to its niche in the human host. Among these are genes encoding two related Type IV pili, the mannose-sensitive haemagglutinin (MSHA) pilus, which aids V. cholerae persistence in aquatic environments but causes clearance of bacteria by host immune defences, and the toxin co-regulated pilus (TCP) required for colonization. These antagonistic effects are resolved transcriptionally by the regulator ToxT, which represses msh genes while activating tcp genes during infection. We show that these two pili systems are also intertwined post-transcriptionally through the ToxT-regulated pre-pilin peptidase TcpJ. We found that the major MSHA pilin, MshA, was degraded in V. cholerae in a TcpJ-dependent fashion. In a heterologous Escherichia coli system, TcpJ can recognize both MshA and its cognate substrate, the TCP subunit TcpA, but that processing by TcpJ causes the degradation of MshA. Through site-directed mutagenesis and chimeric pilin analysis, we show that this process targets a combination of MshA N-terminal motifs and depends on the proteolytic activity of TcpJ. Moreover, overexpression of tcpJ partially restored the ability of bacteria unable to transcriptionally downregulate msh genes to colonize infant mice. These findings describe co-ordinated proteolysis as a regulatory mechanism in V. cholerae and illustrate this organism's adaptability in the face of dramatic environmental changes.

 

 

 

 

 

 

 

 

 

                   

© The Trustees of the University of Pennsylvania