Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.963
Filter
Add more filters

Publication year range
1.
Cell ; 176(1-2): 268-280.e13, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30554875

ABSTRACT

Vibrio cholerae uses a quorum-sensing (QS) system composed of the autoinducer 3,5-dimethylpyrazin-2-ol (DPO) and receptor VqmA (VqmAVc), which together repress genes for virulence and biofilm formation. vqmA genes exist in Vibrio and in one vibriophage, VP882. Phage-encoded VqmA (VqmAPhage) binds to host-produced DPO, launching the phage lysis program via an antirepressor that inactivates the phage repressor by sequestration. The antirepressor interferes with repressors from related phages. Like phage VP882, these phages encode DNA-binding proteins and partner antirepressors, suggesting that they, too, integrate host-derived information into their lysis-lysogeny decisions. VqmAPhage activates the host VqmAVc regulon, whereas VqmAVc cannot induce phage-mediated lysis, suggesting an asymmetry whereby the phage influences host QS while enacting its own lytic-lysogeny program without interference. We reprogram phages to activate lysis in response to user-defined cues. Our work shows that a phage, causing bacterial infections, and V. cholerae, causing human infections, rely on the same signal molecule for pathogenesis.


Subject(s)
Lysogeny/physiology , Pyrazoles/metabolism , Quorum Sensing/physiology , Bacteriophages/metabolism , Biofilms , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Quorum Sensing/genetics , Vibrio/metabolism , Vibrio cholerae/metabolism , Vibrio cholerae/physiology , Virulence , Virus Latency
2.
Cell ; 170(6): 1175-1183.e11, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28867285

ABSTRACT

We serendipitously discovered that the marine bacterium Vibrio fischeri induces sexual reproduction in one of the closest living relatives of animals, the choanoflagellate Salpingoeca rosetta. Although bacteria influence everything from nutrition and metabolism to cell biology and development in eukaryotes, bacterial regulation of eukaryotic mating was unexpected. Here, we show that a single V. fischeri protein, the previously uncharacterized EroS, fully recapitulates the aphrodisiac-like activity of live V. fischeri. EroS is a chondroitin lyase; although its substrate, chondroitin sulfate, was previously thought to be an animal synapomorphy, we demonstrate that S. rosetta produces chondroitin sulfate and thus extend the ancestry of this important glycosaminoglycan to the premetazoan era. Finally, we show that V. fischeri, purified EroS, and other bacterial chondroitin lyases induce S. rosetta mating at environmentally relevant concentrations, suggesting that bacteria likely regulate choanoflagellate mating in nature.


Subject(s)
Aliivibrio fischeri/enzymology , Choanoflagellata/microbiology , Choanoflagellata/physiology , Chondroitinases and Chondroitin Lyases/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Choanoflagellata/cytology , Chondroitin Sulfates/metabolism , Meiosis , Reproduction , Sequence Alignment
3.
Immunity ; 54(8): 1683-1697.e3, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34107298

ABSTRACT

Microbe-derived acetate activates the Drosophila immunodeficiency (IMD) pathway in a subset of enteroendocrine cells (EECs) of the anterior midgut. In these cells, the IMD pathway co-regulates expression of antimicrobial and enteroendocrine peptides including tachykinin, a repressor of intestinal lipid synthesis. To determine whether acetate acts on a cell surface pattern recognition receptor or an intracellular target, we asked whether acetate import was essential for IMD signaling. Mutagenesis and RNA interference revealed that the putative monocarboxylic acid transporter Tarag was essential for enhancement of IMD signaling by dietary acetate. Interference with histone deacetylation in EECs augmented transcription of genes regulated by the steroid hormone ecdysone including IMD targets. Reduced expression of the histone acetyltransferase Tip60 decreased IMD signaling and blocked rescue by dietary acetate and other sources of intracellular acetyl-CoA. Thus, microbe-derived acetate induces chromatin remodeling within enteroendocrine cells, co-regulating host metabolism and intestinal innate immunity via a Tip60-steroid hormone axis that is conserved in mammals.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Enteroendocrine Cells/metabolism , Gastrointestinal Microbiome/immunology , Histone Acetyltransferases/metabolism , Intestines/immunology , Acetates/immunology , Acetyl Coenzyme A/metabolism , Animals , Chromatin Assembly and Disassembly/physiology , Drosophila melanogaster/microbiology , Ecdysone/metabolism , Immunity, Innate/immunology , Intestines/microbiology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , RNA Interference , Signal Transduction/immunology , Tachykinins/metabolism
4.
EMBO J ; 42(3): e111562, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36504455

ABSTRACT

Pandemic and endemic strains of Vibrio cholerae arise from toxigenic conversion by the CTXφ bacteriophage, a process by which CTXφ infects nontoxigenic strains of V. cholerae. CTXφ encodes the cholera toxin, an enterotoxin responsible for the watery diarrhea associated with cholera infections. Despite the critical role of CTXφ during infections, signals that affect CTXφ-driven toxigenic conversion or expression of the CTXφ-encoded cholera toxin remain poorly characterized, particularly in the context of the gut mucosa. Here, we identify mucin polymers as potent regulators of CTXφ-driven pathogenicity in V. cholerae. Our results indicate that mucin-associated O-glycans block toxigenic conversion by CTXφ and suppress the expression of CTXφ-related virulence factors, including the toxin co-regulated pilus and cholera toxin, by interfering with the TcpP/ToxR/ToxT virulence pathway. By synthesizing individual mucin glycan structures de novo, we identify the Core 2 motif as the critical structure governing this virulence attenuation. Overall, our results highlight a novel mechanism by which mucins and their associated O-glycan structures affect CTXφ-mediated evolution and pathogenicity of V. cholerae, underscoring the potential regulatory power housed within mucus.


Subject(s)
Bacteriophages , Cholera Toxin , Mucins , Vibrio cholerae , Virulence , Bacteriophages/genetics , Bacteriophages/pathogenicity , Cholera Toxin/genetics , Cholera Toxin/metabolism , Mucins/genetics , Mucins/metabolism , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Virulence/genetics , Virulence/physiology , Polysaccharides/genetics , Polysaccharides/metabolism
5.
Annu Rev Microbiol ; 76: 681-702, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35759873

ABSTRACT

Cholera is a severe diarrheal disease caused by the bacterium Vibrio cholerae and constitutes a significant public health threat in many areas of the world. V. cholerae infection elicits potent and long-lasting immunity, and efforts to develop cholera vaccines have been ongoing for more than a century. Currently available inactivated two-dose oral cholera vaccines are increasingly deployed to both prevent and actively curb cholera outbreaks, and they are key components of the global effort to eradicate cholera. However, these killed whole-cell vaccines have several limitations, and a variety of new oral and nonoral cholera vaccine platforms have recently been developed. Here, we review emerging concepts in cholera vaccine design and implementation that have been driven by insights from human and animal studies. As a prototypical vaccine-preventable disease, cholera continues to be an excellent target for the development and application of cutting-edge technologies and platforms that may transform vaccinology.


Subject(s)
Cholera Vaccines , Cholera , Vibrio cholerae , Animals , Cholera/prevention & control , Humans , Vaccines, Inactivated
6.
Annu Rev Microbiol ; 76: 503-532, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35671532

ABSTRACT

Biofilms are a widely observed growth mode in which microbial communities are spatially structured and embedded in a polymeric extracellular matrix. Here, we focus on the model bacterium Vibrio cholerae and summarize the current understanding of biofilm formation, including initial attachment, matrix components, community dynamics, social interactions, molecular regulation, and dispersal. The regulatory network that orchestrates the decision to form and disperse from biofilms coordinates various environmental inputs. These cues are integrated by several transcription factors, regulatory RNAs, and second-messenger molecules, including bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP). Through complex mechanisms, V. cholerae weighs the energetic cost of forming biofilms against the benefits of protection and social interaction that biofilms provide.


Subject(s)
Biofilms , Vibrio cholerae , Bacterial Proteins/metabolism , Biofilms/growth & development , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Transcription Factors/metabolism , Vibrio cholerae/genetics , Vibrio cholerae/physiology
7.
Proc Natl Acad Sci U S A ; 121(25): e2316143121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38861595

ABSTRACT

Vibrio vulnificus causes life-threatening wound and gastrointestinal infections, mediated primarily by the production of a Multifunctional-Autoprocessing Repeats-In-Toxin (MARTX) toxin. The most commonly present MARTX effector domain, the Makes Caterpillars Floppy-like (MCF) toxin, is a cysteine protease stimulated by host adenosine diphosphate (ADP) ribosylation factors (ARFs) to autoprocess. Here, we show processed MCF then binds and cleaves host Ras-related proteins in brain (Rab) guanosine triphosphatases within their C-terminal tails resulting in Rab degradation. We demonstrate MCF binds Rabs at the same interface occupied by ARFs. Moreover, we show MCF preferentially binds to ARF1 prior to autoprocessing and is active to cleave Rabs only subsequent to autoprocessing. We then use structure prediction algorithms to demonstrate that structural composition, rather than sequence, determines Rab target specificity. We further determine a crystal structure of aMCF as a swapped dimer, revealing an alternative conformation we suggest represents the open, activated state of MCF with reorganized active site residues. The cleavage of Rabs results in Rab1B dispersal within cells and loss of Rab1B density in the intestinal tissue of infected mice. Collectively, our work describes an extracellular bacterial mechanism whereby MCF is activated by ARFs and subsequently induces the degradation of another small host guanosine triphosphatase (GTPase), Rabs, to drive organelle damage, cell death, and promote pathogenesis of these rapidly fatal infections.


Subject(s)
Bacterial Toxins , Vibrio vulnificus , rab GTP-Binding Proteins , Animals , Female , Humans , Mice , ADP-Ribosylation Factors/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/chemistry , HEK293 Cells , Mice, Inbred ICR , Proteolysis , rab GTP-Binding Proteins/metabolism , Vibrio Infections/microbiology , Vibrio Infections/metabolism , Vibrio vulnificus/metabolism , Vibrio vulnificus/pathogenicity
8.
Annu Rev Microbiol ; 75: 151-174, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34623898

ABSTRACT

Most bacteria are protected from environmental offenses by a cell wall consisting of strong yet elastic peptidoglycan. The cell wall is essential for preserving bacterial morphology and viability, and thus the enzymes involved in the production and turnover of peptidoglycan have become preferred targets for many of our most successful antibiotics. In the past decades, Vibrio cholerae, the gram-negative pathogen causing the diarrheal disease cholera, has become a major model for understanding cell wall genetics, biochemistry, and physiology. More than 100 articles have shed light on novel cell wall genetic determinants, regulatory links, and adaptive mechanisms. Here we provide the first comprehensive review of V. cholerae's cell wall biology and genetics. Special emphasis is placed on the similarities and differences with Escherichia coli, the paradigm for understanding cell wall metabolism and chemical structure in gram-negative bacteria.


Subject(s)
Vibrio cholerae , Biology , Cell Wall/metabolism , Escherichia coli/metabolism , Peptidoglycan/metabolism , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
9.
Proc Natl Acad Sci U S A ; 120(16): e2212664120, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37040409

ABSTRACT

Many bacteria possess dynamic filaments called Type IV pili (T4P) that perform diverse functions in colonization and dissemination, including host cell adhesion, DNA uptake, and secretion of protein substrates-exoproteins-from the periplasm to the extracellular space. The Vibrio cholerae toxin-coregulated pilus (TCP) and the enterotoxigenic Escherichia coli CFA/III pilus each mediates export of a single exoprotein, TcpF and CofJ, respectively. Here, we show that the disordered N-terminal segment of mature TcpF is the export signal (ES) recognized by TCP. Deletion of the ES disrupts secretion and causes TcpF to accumulate in the V. cholerae periplasm. The ES alone can mediate export of Neisseria gonorrhoeae FbpA by V. cholerae in a T4P-dependent manner. The ES is specific for its autologous T4P machinery as CofJ bearing the TcpF ES is exported by V. cholerae, whereas TcpF bearing the CofJ ES is not. Specificity is mediated by binding of the ES to TcpB, a minor pilin that primes pilus assembly and forms a trimer at the pilus tip. Finally, the ES is proteolyzed from the mature TcpF protein upon secretion. Together, these results provide a mechanism for delivery of TcpF across the outer membrane and release into the extracellular space.


Subject(s)
Fimbriae, Bacterial , Vibrio cholerae , Fimbriae, Bacterial/metabolism , Fimbriae Proteins/metabolism , Vibrio cholerae/genetics
10.
Proc Natl Acad Sci U S A ; 120(29): e2304378120, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37428913

ABSTRACT

ToxR, a Vibrio cholerae transmembrane one-component signal transduction factor, lies within a regulatory cascade that results in the expression of ToxT, toxin coregulated pilus, and cholera toxin. While ToxR has been extensively studied for its ability to activate or repress various genes in V. cholerae, here we present the crystal structures of the ToxR cytoplasmic domain bound to DNA at the toxT and ompU promoters. The structures confirm some predicted interactions, yet reveal other unexpected promoter interactions with implications for other potential regulatory roles for ToxR. We show that ToxR is a versatile virulence regulator that recognizes diverse and extensive, eukaryotic-like regulatory DNA sequences, that relies more on DNA structural elements than specific sequences for binding. Using this topological DNA recognition mechanism, ToxR can bind both in tandem and in a twofold inverted-repeat-driven manner. Its regulatory action is based on coordinated multiple binding to promoter regions near the transcription start site, which can remove the repressing H-NS proteins and prepares the DNA for optimal interaction with the RNA polymerase.


Subject(s)
Vibrio cholerae , Vibrio cholerae/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Virulence , Bacterial Proteins/metabolism , DNA/genetics , DNA/metabolism , Gene Expression Regulation, Bacterial
11.
Mol Microbiol ; 121(5): 850-864, 2024 05.
Article in English | MEDLINE | ID: mdl-38323722

ABSTRACT

The diarrheal disease cholera is caused by the versatile and responsive bacterium Vibrio cholerae, which is capable of adapting to environmental changes. Among others, the alternative sigma factor RpoS activates response pathways, including regulation of motility- and chemotaxis-related genes under nutrient-poor conditions in V. cholerae. Although RpoS has been well characterised, links between RpoS and other regulatory networks remain unclear. In this study, we identified the ArcAB two-component system to control rpoS transcription and RpoS protein stability in V. cholerae. In a manner similar to that seen in Escherichia coli, the ArcB kinase not only activates the response regulator ArcA but also RssB, the anti-sigma factor of RpoS. Our results demonstrated that, in V. cholerae, RssB is phosphorylated by ArcB, which subsequently activates RpoS proteolysis. Furthermore, ArcA acts as a repressor of rpoS transcription. Additionally, we determined that the cysteine residue at position 180 of ArcB is crucial for signal recognition and activity. Thus, our findings provide evidence linking RpoS response to the anoxic redox control system ArcAB in V. cholerae.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Sigma Factor , Vibrio cholerae , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chemotaxis/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Regulatory Networks , Phosphorylation , Repressor Proteins/metabolism , Repressor Proteins/genetics , Sigma Factor/metabolism , Sigma Factor/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
12.
EMBO J ; 40(24): e108542, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34612526

ABSTRACT

Bacterial small RNAs (sRNAs) are well known to modulate gene expression by base pairing with trans-encoded transcripts and are typically non-coding. However, several sRNAs have been reported to also contain an open reading frame and thus are considered dual-function RNAs. In this study, we discovered a dual-function RNA from Vibrio cholerae, called VcdRP, harboring a 29 amino acid small protein (VcdP), as well as a base-pairing sequence. Using a forward genetic screen, we identified VcdRP as a repressor of cholera toxin production and link this phenotype to the inhibition of carbon transport by the base-pairing segment of the regulator. By contrast, we demonstrate that the VcdP small protein acts downstream of carbon transport by binding to citrate synthase (GltA), the first enzyme of the citric acid cycle. Interaction of VcdP with GltA results in increased enzyme activity and together VcdR and VcdP reroute carbon metabolism. We further show that transcription of vcdRP is repressed by CRP allowing us to provide a model in which VcdRP employs two different molecular mechanisms to synchronize central metabolism in V. cholerae.


Subject(s)
Carbon/metabolism , Cholera Toxin/metabolism , Citrate (si)-Synthase/metabolism , RNA, Bacterial/genetics , Vibrio cholerae/metabolism , Bacterial Proteins/metabolism , Biological Transport , Down-Regulation , Gene Expression Regulation, Bacterial , Genetic Testing , Open Reading Frames , Phenotype , RNA, Bacterial/metabolism , Vibrio cholerae/genetics
13.
Proc Natl Acad Sci U S A ; 119(11): e2121180119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35254905

ABSTRACT

SignificanceIn a polymicrobial battlefield where different species compete for nutrients and colonization niches, antimicrobial compounds are the sword and shield of commensal microbes in competition with invading pathogens and each other. The identification of an Escherichia coli-produced genotoxin, colibactin, and its specific targeted killing of enteric pathogens and commensals, including Vibrio cholerae and Bacteroides fragilis, sheds light on our understanding of intermicrobial interactions in the mammalian gut. Our findings elucidate the mechanisms through which genotoxins shape microbial communities and provide a platform for probing the larger role of enteric multibacterial interactions regarding infection and disease outcomes.


Subject(s)
Cholera/microbiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Microbial Interactions , Mutagens/metabolism , Vibrio cholerae/physiology , Animals , Antibiosis , Cholera/mortality , DNA Damage , Disease Models, Animal , Escherichia coli/physiology , Humans , Mice , Peptides/metabolism , Peptides/pharmacology , Polyketides/metabolism , Polyketides/pharmacology , Prognosis , Reactive Oxygen Species , Vibrio cholerae/drug effects
14.
Proc Natl Acad Sci U S A ; 119(46): e2210115119, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36343262

ABSTRACT

Although the mechanism by which the cyclic AMP receptor protein (CRP) regulates global gene transcription has been intensively studied for decades, new discoveries remain to be made. Here, we report that, during rapid growth, CRP associates with both the well-conserved, dual-function DNA-binding protein peptidase A (PepA) and the cell membrane. These interactions are not present under nutrient-limited growth conditions, due to post-translational modification of three lysines on a single face of CRP. Although coincident DNA binding is rare, dissociation from CRP results in increased PepA occupancy at many chromosomal binding sites and differential regulation of hundreds of genes, including several encoding cyclic dinucleotide phosphodiesterases. We show that PepA represses biofilm formation and activates motility/chemotaxis. We propose a model in which membrane-bound CRP interferes with PepA DNA binding. Under nutrient limitation, PepA is released. Together, CRP and free PepA activate a transcriptional response that impels the bacterium to seek a more hospitable environment. This work uncovers a function for CRP in the sequestration of a regulatory protein. More broadly, it describes a paradigm of bacterial transcriptome modulation through metabolically regulated association of transcription factors with the cell membrane.


Subject(s)
Cyclic AMP Receptor Protein , Vibrio cholerae , Cyclic AMP Receptor Protein/genetics , Cyclic AMP Receptor Protein/metabolism , Vibrio cholerae/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transcription Factors/metabolism , DNA/metabolism
15.
J Bacteriol ; 206(5): e0003524, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38695522

ABSTRACT

Vibrio (Aliivibrio) fischeri's initial rise to fame derived from its alluring production of blue-green light. Subsequent studies to probe the mechanisms underlying this bioluminescence helped the field discover the phenomenon now known as quorum sensing. Orthologs of quorum-sensing regulators (i.e., LuxR and LuxI) originally identified in V. fischeri were subsequently uncovered in a plethora of bacterial species, and analogous pathways were found in yet others. Over the past three decades, the study of this microbe has greatly expanded to probe the unique role of V. fischeri as the exclusive symbiont of the light organ of the Hawaiian bobtail squid, Euprymna scolopes. Buoyed by this optically amenable host and by persistent and insightful researchers who have applied novel and cross-disciplinary approaches, V. fischeri has developed into a robust model for microbe-host associations. It has contributed to our understanding of how bacteria experience and respond to specific, often fluxing environmental conditions and the mechanisms by which bacteria impact the development of their host. It has also deepened our understanding of numerous microbial processes such as motility and chemotaxis, biofilm formation and dispersal, and bacterial competition, and of the relevance of specific bacterial genes in the context of colonizing an animal host. Parallels in these processes between this symbiont and bacteria studied as pathogens are readily apparent, demonstrating functional conservation across diverse associations and permitting a reinterpretation of "pathogenesis." Collectively, these advances built a foundation for microbiome studies and have positioned V. fischeri to continue to expand the frontiers of our understanding of the microbial world inside animals.


Subject(s)
Aliivibrio fischeri , Decapodiformes , Quorum Sensing , Aliivibrio fischeri/genetics , Aliivibrio fischeri/physiology , Animals , Decapodiformes/microbiology , Symbiosis , Gene Expression Regulation, Bacterial , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Host Microbial Interactions
16.
J Bacteriol ; : e0014324, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39230524

ABSTRACT

A major challenge faced by Vibrio cholerae is constant predation by bacteriophage (phage) in aquatic reservoirs and during infection of human hosts. To overcome phage predation, V. cholerae has acquired and/or evolved a myriad of phage defense systems. Although several novel defense systems have been discovered, we hypothesized that more were encoded in V. cholerae given the low diversity of phages that have been isolated, which infect this species. Using a V. cholerae genomic library, we identified a Type IV restriction system consisting of two genes within a 16-kB region of the Vibrio pathogenicity island-2, which we name TgvA and TgvB (Type I-embedded gmrSD-like system of VPI-2). We show that both TgvA and TgvB are required for defense against T2, T4, and T6 by targeting glucosylated 5-hydroxymethylcytosine (5hmC). T2 or T4 phages that lose the glucose modifications are resistant to TgvAB defense but exhibit a significant evolutionary tradeoff, becoming susceptible to other Type IV restriction systems that target unglucosylated 5hmC. We also show that the Type I restriction-modification system that embeds the tgvAB genes protects against phage T3, secΦ18, secΦ27, and λ, suggesting that this region is a phage defense island. Our study uncovers a novel Type IV restriction system in V. cholerae, increasing our understanding of the evolution and ecology of V. cholerae, while highlighting the evolutionary interplay between restriction systems and phage genome modification.IMPORTANCEBacteria are constantly being predated by bacteriophage (phage). To counteract this predation, bacteria have evolved a myriad of defense systems. Some of these systems specifically digest infecting phage by recognizing unique base modifications present on the phage DNA. In this study, we discover a Type IV restriction system encoded in V. cholerae, which we name TgvAB, and demonstrate it recognizes and restricts phage that have 5-hydroxymethylcytosine glucosylated DNA. Moreover, the evolution of resistance to TgvAB render phage susceptible to other Type IV restriction systems, demonstrating a significant evolutionary tradeoff. These results enhance our understanding of the evolution of V. cholerae and more broadly how bacteria evade phage predation.

17.
J Bacteriol ; 206(2): e0037023, 2024 02 22.
Article in English | MEDLINE | ID: mdl-38270381

ABSTRACT

Multicellular communities of adherent bacteria known as biofilms are often detrimental in the context of a human host, making it important to study their formation and dispersal, especially in animal models. One such model is the symbiosis between the squid Euprymna scolopes and the bacterium Vibrio fischeri. Juvenile squid hatch aposymbiotically and selectively acquire their symbiont from natural seawater containing diverse environmental microbes. Successful pairing is facilitated by ciliary movements that direct bacteria to quiet zones on the surface of the squid's symbiotic light organ where V. fischeri forms a small aggregate or biofilm. Subsequently, the bacteria disperse from that aggregate to enter the organ, ultimately reaching and colonizing deep crypt spaces. Although transient, aggregate formation is critical for optimal colonization and is tightly controlled. In vitro studies have identified a variety of polysaccharides and proteins that comprise the extracellular matrix. Some of the most well-characterized matrix factors include the symbiosis polysaccharide (SYP), cellulose polysaccharide, and LapV adhesin. In this review, we discuss these components, their regulation, and other less understood V. fischeri biofilm contributors. We also highlight what is currently known about dispersal from these aggregates and host cues that may promote it. Finally, we briefly describe discoveries gleaned from the study of other V. fischeri isolates. By unraveling the complexities involved in V. fischeri's control over matrix components, we may begin to understand how the host environment triggers transient biofilm formation and dispersal to promote this unique symbiotic relationship.


Subject(s)
Aliivibrio fischeri , Biofilms , Animals , Humans , Aliivibrio fischeri/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Adhesins, Bacterial , Decapodiformes/microbiology , Symbiosis , Polysaccharides
18.
J Bacteriol ; 206(4): e0043323, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38493438

ABSTRACT

In bacteria, disulfide bonds contribute to the folding and stability of proteins important for processes in the cellular envelope. In Escherichia coli, disulfide bond formation is catalyzed by DsbA and DsbB enzymes. DsbA is a periplasmic protein that catalyzes disulfide bond formation in substrate proteins, while DsbB is an inner membrane protein that transfers electrons from DsbA to quinones, thereby regenerating the DsbA active state. Actinobacteria including mycobacteria use an alternative enzyme named VKOR, which performs the same function as DsbB. Disulfide bond formation enzymes, DsbA and DsbB/VKOR, represent novel drug targets because their inhibition could simultaneously affect the folding of several cell envelope proteins including virulence factors, proteins involved in outer membrane biogenesis, cell division, and antibiotic resistance. We have previously developed a cell-based and target-based assay to identify molecules that inhibit the DsbB and VKOR in pathogenic bacteria, using E. coli cells expressing a periplasmic ß-Galactosidase sensor (ß-Galdbs), which is only active when disulfide bond formation is inhibited. Here, we report the construction of plasmids that allows fine-tuning of the expression of the ß-Galdbs sensor and can be mobilized into other gram-negative organisms. As an example, when expressed in Pseudomonas aeruginosa UCBPP-PA14, which harbors two DsbB homologs, ß-Galdbs behaves similarly as in E. coli, and the biosensor responds to the inhibition of the two DsbB proteins. Thus, these ß-Galdbs reporter plasmids provide a basis to identify novel inhibitors of DsbA and DsbB/VKOR in multidrug-resistant gram-negative pathogens and to further study oxidative protein folding in diverse gram-negative bacteria. IMPORTANCE: Disulfide bonds contribute to the folding and stability of proteins in the bacterial cell envelope. Disulfide bond-forming enzymes represent new drug targets against multidrug-resistant bacteria because inactivation of this process would simultaneously affect several proteins in the cell envelope, including virulence factors, toxins, proteins involved in outer membrane biogenesis, cell division, and antibiotic resistance. Identifying the enzymes involved in disulfide bond formation in gram-negative pathogens as well as their inhibitors can contribute to the much-needed antibacterial innovation. In this work, we developed sensors of disulfide bond formation for gram-negative bacteria. These tools will enable the study of disulfide bond formation and the identification of inhibitors for this crucial process in diverse gram-negative pathogens.


Subject(s)
Bacterial Proteins , Escherichia coli , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Virulence Factors/metabolism , Disulfides/chemistry , Oxidation-Reduction
19.
J Proteome Res ; 23(7): 2397-2407, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38904328

ABSTRACT

Protein succinylation modification is a common post-translational modification (PTM) that plays an important role in bacterial metabolic regulation. In this study, quantitative analysis was conducted on the succinylated proteome of wild-type and florfenicol-resistant Vibrio alginolyticus to investigate the mechanism of succinylation regulating antibiotic resistance. Bioinformatic analysis showed that the differentially succinylated proteins were mainly enriched in energy metabolism, and it was found that the succinylation level of phosphoenolpyruvate carboxyl kinase (PEPCK) was highly expressed in the florfenicol-resistant strain. Site-directed mutagenesis was used to mutate the lysine (K) at the succinylation site of PEPCK to glutamic acid (E) and arginine (R), respectively, to investigate the function of lysine succinylation of PEPCK in the florfenicol resistance of V. alginolyticus. The detection of site-directed mutagenesis strain viability under florfenicol revealed that the survival rate of the E mutant was significantly higher than that of the R mutant and wild type, indicating that succinylation modification of PEPCK protein may affect the resistance of V. alginolyticus to florfenicol. This study indicates the important role of PEPCK during V. alginolyticus antibiotic-resistance evolution and provides a theoretical basis for the prevention and control of vibriosis and the development of new antibiotics.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Lysine , Protein Processing, Post-Translational , Thiamphenicol , Vibrio alginolyticus , Thiamphenicol/pharmacology , Thiamphenicol/analogs & derivatives , Thiamphenicol/metabolism , Vibrio alginolyticus/genetics , Vibrio alginolyticus/drug effects , Vibrio alginolyticus/metabolism , Drug Resistance, Bacterial/genetics , Lysine/metabolism , Anti-Bacterial Agents/pharmacology , Mutagenesis, Site-Directed , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Succinic Acid/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics
20.
J Biol Chem ; 299(12): 105386, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37898401

ABSTRACT

Aggregation behavior provides bacteria protection from harsh environments and threats to survival. Two uncharacterized proteases, LapX and Lap, are important for Vibrio cholerae liquid-based aggregation. Here, we determined that LapX is a serine protease with a preference for cleavage after glutamate and glutamine residues in the P1 position, which processes a physiologically based peptide substrate with a catalytic efficiency of 180 ± 80 M-1s-1. The activity with a LapX substrate identified by a multiplex substrate profiling by mass spectrometry screen was 590 ± 20 M-1s-1. Lap shares high sequence identity with an aminopeptidase (termed VpAP) from Vibrio proteolyticus and contains an inhibitory bacterial prepeptidase C-terminal domain that, when eliminated, increases catalytic efficiency on leucine p-nitroanilide nearly four-fold from 5.4 ± 4.1 × 104 M-1s-1 to 20.3 ± 4.3 × 104 M-1s-1. We demonstrate that LapX processes Lap to its mature form and thus amplifies Lap activity. The increase is approximately eighteen-fold for full-length Lap (95.7 ± 5.6 × 104 M-1s-1) and six-fold for Lap lacking the prepeptidase C-terminal domain (11.3 ± 1.9 × 105 M-1s-1). In addition, substrate profiling reveals preferences for these two proteases that could inform in vivo function. Furthermore, purified LapX and Lap restore the timing of the V. cholerae aggregation program to a mutant lacking the lapX and lap genes. Both proteases must be present to restore WT timing, and thus they appear to act sequentially: LapX acts on Lap, and Lap acts on the substrate involved in aggregation.


Subject(s)
Bacterial Proteins , Leucyl Aminopeptidase , Serine Proteases , Vibrio cholerae , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Leucyl Aminopeptidase/chemistry , Leucyl Aminopeptidase/genetics , Leucyl Aminopeptidase/physiology , Peptides , Serine Proteases/chemistry , Serine Proteases/genetics , Serine Proteases/physiology , Substrate Specificity , Vibrio cholerae/enzymology , Vibrio cholerae/genetics , Vibrio cholerae/physiology , Catalysis
SELECTION OF CITATIONS
SEARCH DETAIL