Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Metab ; 3(8): 1071-1090, 2021 08.
Article in English | MEDLINE | ID: mdl-34341568

ABSTRACT

Metabolic health depends on the brain's ability to control food intake and nutrient use versus storage, processes that require peripheral signals such as the adipocyte-derived hormone, leptin, to cross brain barriers and mobilize regulatory circuits. We have previously shown that hypothalamic tanycytes shuttle leptin into the brain to reach target neurons. Here, using multiple complementary models, we show that tanycytes express functional leptin receptor (LepR), respond to leptin by triggering Ca2+ waves and target protein phosphorylation, and that their transcytotic transport of leptin requires the activation of a LepR-EGFR complex by leptin and EGF sequentially. Selective deletion of LepR in tanycytes blocks leptin entry into the brain, inducing not only increased food intake and lipogenesis but also glucose intolerance through attenuated insulin secretion by pancreatic ß-cells, possibly via altered sympathetic nervous tone. Tanycytic LepRb-EGFR-mediated transport of leptin could thus be crucial to the pathophysiology of diabetes in addition to obesity, with therapeutic implications.


Subject(s)
Brain/metabolism , Ependymoglial Cells/metabolism , ErbB Receptors/metabolism , Leptin/metabolism , Lipid Metabolism , Pancreas/metabolism , Receptors, Leptin/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Energy Metabolism , Insulin-Secreting Cells/metabolism , Phosphorylation
2.
Endocrinology ; 162(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34388249

ABSTRACT

The brain influences liver metabolism through many neuroendocrine and autonomic mechanisms that have evolved to protect the organism against starvation and hypoglycemia. Unfortunately, this effective way of preventing death has become dysregulated in modern obesogenic environments, although the pathophysiological mechanisms behind metabolic dyshomeostasis are still unclear. In this Mini-Review, we provide our thoughts regarding obesity and type 2 diabetes as diseases of the autonomic nervous system. We discuss the pathophysiological mechanisms that alter the autonomic brain-liver communication in these diseases, and how they could represent important targets to prevent or treat metabolic dysfunctions. We discuss how sympathetic hyperactivity to the liver may represent an early event in the progression of metabolic diseases and could progressively lead to hepatic neuropathy. We hope that this discussion will inspire and help to frame a model based on better understanding of the chronology of autonomic dysfunctions in the liver, enabling the application of the right strategy at the right time.


Subject(s)
Autonomic Nervous System/physiopathology , Brain/physiopathology , Liver/physiopathology , Metabolic Diseases , Animals , Autonomic Nervous System/pathology , Brain/metabolism , Cell Communication , Humans , Liver/innervation , Liver/metabolism , Metabolic Diseases/physiopathology , Metabolic Diseases/psychology , Neurosecretory Systems/physiopathology
3.
Neuroendocrinology ; 111(4): 370-387, 2021.
Article in English | MEDLINE | ID: mdl-32335558

ABSTRACT

INTRODUCTION: Metabolic dysfunction is now recognized as a pivotal component of Alzheimer's disease (AD), the most common dementia worldwide. However, the precise molecular mechanisms linking metabolic dysfunction to AD remain elusive. OBJECTIVE: Here, we investigated the direct impact of soluble oligomeric amyloid beta (Aß) peptides, the main molecular hallmark of AD, on the leptin system, a major component of central energy metabolism regulation. METHODS: We developed a new time-resolved fluorescence resonance energy transfer-based Aß binding assay for the leptin receptor (LepR) and studied the effect of Aß on LepR function in several in vitro assays. The in vivo effect of Aß on LepR function was studied in an Aß-specific AD mouse model and in pro-opiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus. RESULTS: We revealed specific and high-affinity (Ki = 0.1 nM) binding of Aß to LepR. Pharmacological characterization of this interaction showed that Aß binds allosterically to the extracellular domain of LepR and negatively affects receptor function. Negative allosteric modulation of LepR by Aß was detected at the level of signaling pathways (STAT-3, AKT, and ERK) in vitroand in vivo. Importantly, the leptin-induced response of POMC neurons, key players in the regulation of metabolic function, was completely abolished in the presence of Aß. CONCLUSION: Our data indicate that Aß is a negative allosteric modulator of LepR, resulting in impaired leptin action, and qualify LepR as a new and direct target of Aß oligomers. Preventing the interaction of Aß with LepR might improve both the metabolic and cognitive dysfunctions in AD condition.


Subject(s)
Allosteric Regulation/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Leptin/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, Leptin/metabolism , Animals , Cell Line , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice , Signal Transduction/physiology
4.
J Microbiol Methods ; 165: 105702, 2019 10.
Article in English | MEDLINE | ID: mdl-31454505

ABSTRACT

We describe a proteomic approach to identify transcription factors binding to a target promoter. The method's usefulness was tested by identifying proteins binding to the Vibrio cholerae rpoS promoter in response to cell density. Proteins identified in this screen included the nucleoid-associated protein Fis and the quorum sensing regulator HapR.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Sigma Factor/genetics , Transcription Factors/genetics , Vibrio cholerae/genetics , Promoter Regions, Genetic , Proteome/metabolism , Quorum Sensing , Transcription, Genetic
5.
Cell Mol Life Sci ; 76(6): 1201-1214, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30659329

ABSTRACT

Leptin links body energy stores to high energy demanding processes like reproduction and immunity. Based on leptin's role in autoimmune diseases and cancer, several leptin and leptin receptor (LR) antagonists have been developed, but these intrinsically lead to unwanted weight gain. Here, we report on the uncoupling of leptin's metabolic and immune functions based on the cross talk with the epidermal growth factor receptor (EGFR). We show that both receptors spontaneously interact and, remarkably, that this complex can partially overrule the lack of LR activation by a leptin antagonistic mutein. Moreover, this leptin mutant induces EGFR phosphorylation comparable to wild-type leptin. Exploiting this non-canonical leptin signalling pathway, we identified a camelid single-domain antibody that selectively inhibits this LR-EGFR cross talk without interfering with homotypic LR signalling. Administration in vivo showed that this single-domain antibody did not interfere with leptin's metabolic functions, but could reverse the leptin-driven protection against starvation-induced thymic and splenic atrophy. These findings offer new opportunities for the design and clinical application of selective leptin and LR antagonists that avoid unwanted metabolic side effects.


Subject(s)
Leptin/immunology , Leptin/metabolism , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/metabolism , Single-Domain Antibodies/pharmacology , Animals , Camelids, New World/immunology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Leptin/genetics , Ligands , Mice, Inbred C57BL , Mutation , Protein Binding/drug effects , Receptor Cross-Talk/drug effects , Receptors, Leptin/genetics , Signal Transduction
6.
Methods Mol Biol ; 1839: 65-75, 2018.
Article in English | MEDLINE | ID: mdl-30047055

ABSTRACT

Chromatin immunoprecipitation (ChIP) measures the physical association between a protein and DNA in the cell. In combination with next-generation sequencing, the technique enables the identification of DNA targets for the corresponding protein across an entire genome. Here we describe the immunoprecipitation of Vibrio cholerae DNA bound to the histone-like nucleoid structuring protein (H-NS) tagged with the Flag epitope. The quality of the DNA obtained in this protocol is suitable for next-generation sequencing. The procedure described herein can be readily adapted to other bacteria and DNA-binding proteins.


Subject(s)
Chromatin Immunoprecipitation , Bacterial Proteins/metabolism , DNA-Binding Proteins , High-Throughput Nucleotide Sequencing , Protein Binding , Real-Time Polymerase Chain Reaction , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
7.
Microbiology (Reading) ; 164(7): 998-1003, 2018 07.
Article in English | MEDLINE | ID: mdl-29813015

ABSTRACT

Hypervirulent atypical El Tor biotype Vibrio cholerae O1 isolates harbour mutations in the DNA-binding domain of the nucleoid-associated protein H-NS and the receiver domain of the response regulator VieA. Here, we provide two examples in which inactivation of H-NS in El Tor biotype vibrios unmasks hidden regulatory connections. First, deletion of the helix-turn-helix domain of VieA in an hns mutant background diminished biofilm formation and exopolysaccharide gene expression, a function that phenotypically opposes its phosphodiesterase activity. Second, deletion of vieA in an hns mutant diminished the expression of σE, a virulence determinant that mediates the envelope stress response. hns mutants were highly sensitive to envelope stressors compared to wild-type. However, deletion of vieA in the hns mutant restored or exceeded wild-type resistance. These findings suggest an evolutionary path for the emergence of hypervirulent strains starting from nucleotide sequence diversification affecting the interaction of H-NS with DNA.


Subject(s)
Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Bacterial , Vibrio cholerae O1/genetics , Vibrio cholerae O1/pathogenicity , Biofilms/growth & development , Gene Deletion , Mutation , Polysaccharides, Bacterial/genetics , Sigma Factor/genetics , Stress, Physiological/genetics , Vibrio cholerae O1/physiology , Virulence/genetics
8.
Mol Microbiol ; 107(3): 330-343, 2018 02.
Article in English | MEDLINE | ID: mdl-29152799

ABSTRACT

VieA is a cyclic diguanylate phosphodiesterase that modulates biofilm development and motility in Vibrio cholerae O1 of the classical biotype. vieA is part of an operon encoding the VieSAB signal transduction pathway that is nearly silent in V. cholerae of the El Tor biotype. A DNA pull-down assay for proteins interacting with the vieSAB promoter identified the LysR-type regulator LeuO. We show that in classical biotype V. cholerae, LeuO cooperates with the nucleoid-associated protein H-NS to repress vieSAB transcription. LeuO and H-NS interacted with the vieSAB promoter of both biotypes with similar affinities and protected overlapping DNA sequences. H-NS was expressed at similar levels in both cholera biotypes. In contrast, El Tor biotype strains expressed negligible LeuO under identical conditions. In El Tor biotype vibrios, transcription of vieSAB is repressed by the quorum sensing regulator HapR, which is absent in classical biotype strains. Restoring HapR expression in classical biotype V. cholerae repressed vieSAB transcription by binding to its promoter. We propose that double locking of the vieSAB promoter by H-NS and HapR in the El Tor biotype prior to the cessation of exponential growth results in a more pronounced decline in VieA specific activity compared to the classical biotype.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Operon/genetics , Promoter Regions, Genetic/genetics , Quorum Sensing/genetics , Transcription Factors/metabolism , Transcription, Genetic , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Vibrio cholerae O1/genetics , Vibrio cholerae O1/metabolism , Virulence/genetics
9.
Microb Pathog ; 113: 17-24, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29038053

ABSTRACT

Vibrio cholerae of serogroups O1 and O139, the causative agent of Asiatic cholera, continues to be a major global health threat. This pathogen utilizes substratum-specific pili to attach to distinct surfaces in the aquatic environment and the human small intestine and detaches when conditions become unfavorable. Both attachment and detachment are critical to bacterial environmental survival, pathogenesis and disease transmission. However, the factors that promote detachment are less understood. In this study, we examine the role of flagellar motility and hemagglutinin/protease (HapA) in vibrio detachment from a non-degradable abiotic surface and from the suckling mouse intestine. Flagellar motility facilitated V. cholerae detachment from abiotic surfaces. HapA had no effect on the stability of biofilms formed on abiotic surfaces despite representing >50% of the proteolytic activity present in the extracellular matrix. We developed a balanced lethal plasmid system to increase the bacterial cyclic diguanylate (c-di-GMP) pool late in infection, a condition that represses motility and HapA expression. Increasing the c-di-GMP pool enhanced V. cholerae colonization of the suckling mouse intestine. The c-di-GMP effect was fully abolished in hapA isogenic mutants. These results suggest that motility facilitates detachment in a substratum-independent manner. Instead, HapA appears to function as a substratum-specific detachment factor.


Subject(s)
Bacterial Adhesion/physiology , Biofilms/growth & development , Flagella/physiology , Intestinal Mucosa/microbiology , Metalloendopeptidases/metabolism , Movement/physiology , Vibrio cholerae/metabolism , Animals , Cholera/microbiology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Fimbriae, Bacterial/physiology , Gene Expression Regulation, Bacterial , Intestine, Small/microbiology , Metalloendopeptidases/genetics , Mice , Polystyrenes , Vibrio cholerae/genetics
10.
Microbes Infect ; 19(6): 370-375, 2017 06.
Article in English | MEDLINE | ID: mdl-28392408

ABSTRACT

A screen for inhibitors of Vibrio cholerae motility identified the compound 3-amino 1,8-naphthalimide (3-A18NI), a structural analog of the cholera drug virstatin. Similar to virstatin, 3-A18NI diminished cholera toxin production. In contrast, 3-A18NI impeded swimming and/or swarming motility of V. cholerae and V. parahemolyticus suggesting that it could target the chemotaxis pathway shared by the polar and lateral flagellar system of vibrios. 3-A18NI did not inhibit the expression of V. cholerae major flagellin FlaA or the assembly of its polar flagellum. Finally, 3-A18NI enhanced V. cholerae colonization mimicking the phenotype of chemotaxis mutants that exhibit counterclockwise-biased flagellum rotation.


Subject(s)
1-Naphthylamine/analogs & derivatives , Butyrates/pharmacology , Cholera/drug therapy , Naphthalimides/pharmacology , Quinolones/pharmacology , Vibrio cholerae/drug effects , 1-Naphthylamine/pharmacology , Animals , Bacterial Proteins/metabolism , Cholera Toxin/biosynthesis , Flagella/drug effects , Flagella/physiology , Flagellin/metabolism , Mice , Vibrio cholerae/physiology
11.
Res Microbiol ; 168(1): 16-25, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27492955

ABSTRACT

Vibrio cholerae has become a model organism for studies connecting virulence, pathogen evolution and infectious disease ecology. The coordinate expression of motility, virulence and biofilm enhances its pathogenicity, environmental fitness and fecal-oral transmission. The histone-like nucleoid structuring protein negatively regulates gene expression at multiple phases of the V. cholerae life cycle. Here we discuss: (i) the regulatory and structural implications of H-NS chromatin-binding in the two-chromosome cholera bacterium; (ii) the factors that counteract H-NS repression; and (iii) a model for the regulation of the V. cholerae life cycle that integrates H-NS repression, cyclic diguanylic acid signaling and the general stress response.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Vibrio cholerae/genetics , Vibrio cholerae/physiology , Biofilms/growth & development , Chromatin/metabolism , Locomotion , Protein Binding , Virulence
12.
Toxicon ; 115: 55-62, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26952544

ABSTRACT

Vibrio cholerae of serogroup O1 and O139, the etiological agent of the diarrheal disease cholera, expresses the extracellular Zn-dependent metalloprotease hemagglutinin (HA)/protease also reported as vibriolysin. This enzyme is also produced by non-O1/O139 (non-cholera) strains that cause mild, sporadic illness (i.e. gastroenteritis, wound or ear infections). Orthologs of HA/protease are present in other members of the Vibrionaceae family pathogenic to humans and fish. HA/protease belongs to the M4 neutral peptidase family and displays significant amino acid sequence homology to Pseudomonas aeruginosa elastase (LasB) and Bacillus thermoproteolyticus thermolysin. It exhibits a broad range of potentially pathogenic activities in cell culture and animal models. These activities range from the covalent modification of other toxins, the degradation of the protective mucus barrier and disruption of intestinal tight junctions. Here we review (i) the structure and regulation of HA/protease expression, (ii) its interaction with other toxins and the intestinal mucosa and (iii) discuss the possible role(s) of HA/protease in the pathogenesis of cholera.


Subject(s)
Bacterial Proteins/chemistry , Metalloendopeptidases/chemistry , Vibrio cholerae/enzymology , Amino Acid Sequence , Animals , Cholera/microbiology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Models, Animal , Sequence Homology, Amino Acid , Thermolysin/chemistry , Vibrio cholerae/genetics
13.
PLoS Negl Trop Dis ; 10(2): e0004330, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26845681

ABSTRACT

Vibrio cholerae can switch between motile and biofilm lifestyles. The last decades have been marked by a remarkable increase in our knowledge of the structure, regulation, and function of biofilms formed under laboratory conditions. Evidence has grown suggesting that V. cholerae can form biofilm-like aggregates during infection that could play a critical role in pathogenesis and disease transmission. However, the structure and regulation of biofilms formed during infection, as well as their role in intestinal colonization and virulence, remains poorly understood. Here, we review (i) the evidence for biofilm formation during infection, (ii) the coordinate regulation of biofilm and virulence gene expression, and (iii) the host signals that favor V. cholerae transitions between alternative lifestyles during intestinal colonization, and (iv) we discuss a model for the role of V. cholerae biofilms in pathogenicity.


Subject(s)
Biofilms/growth & development , Cholera/microbiology , Gastrointestinal Tract/microbiology , Vibrio cholerae/physiology , Virulence Factors/biosynthesis , Humans , Virulence
14.
Genom Data ; 5: 147-150, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26097806

ABSTRACT

The data described in this article pertain to the genome-wide transcription profiling of a Vibrio cholerae mutant lacking the histone-like nucleoid structuring protein (H-NS) and the mapping of the H-NS chromosome binding sites [1, 2]. H-NS is a nucleoid-associated protein with two interrelated functions: organization of the bacterial nucleoid and transcriptional silencing [3]. Both functions require DNA binding and protein oligomerization [4, 5]. H-NS commonly silences the expression of virulence factors acquired by lateral gene transfer [6]. The highly pleiotropic nature of hns mutants in V. cholerae indicates that H-NS impacts a broad range of cellular processes such as virulence, stress response, surface attachment, biofilm development, motility and chemotaxis. We used a V. cholerae strain harboring a deletion of hns and a strain expressing H-NS tagged at the C-terminus with the FLAG epitope to generate datasets representing the hns transcriptome and DNA binding profile under laboratory conditions (LB medium, 37°C). The datasets are publicly available at the Gene Expression Omnibus (GEO) repository (http://www.ncbi.nlm.nih.gov/geo/) with accession numbers GSE62785 and GSE64249.

15.
Mol Microbiol ; 97(4): 630-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25982817

ABSTRACT

Expression of Vibrio cholerae genes required for the biosynthesis of exopolysacchide (vps) and protein (rbm) components of the biofilm matrix is enhanced by cyclic diguanylate (c-di-GMP). In a previous study, we reported that the histone-like nucleoid structuring (H-NS) protein represses the transcription of vpsA, vpsL and vpsT. Here we demonstrate that the regulator VpsT can disrupt repressive H-NS nucleoprotein complexes at the vpsA and vpsL promoters in the presence of c-di-GMP, while H-NS could disrupt the VpsT-promoter complexes in the absence of c-di-GMP. Chromatin immunoprecipitation-Seq showed a remarkable trend for H-NS to cluster at loci involved in biofilm development such as the rbmABCDEF genes. We show that the antagonistic relationship between VpsT and H-NS regulates the expression of the rbmABCDEF cluster. Epistasis analysis demonstrated that VpsT functions as an antirepressor at the rbmA/F, vpsU and vpsA/L promoters. Deletion of vpsT increased H-NS occupancy at these promoters while increasing the c-di-GMP pool had the opposite effect and included the vpsT promoter. The negative effect of c-di-GMP on H-NS occupancy at the vpsT promoter required the regulator VpsR. These results demonstrate that c-di-GMP activates the transcription of genes required for the biosynthesis of the biofilm matrix by triggering a coordinated VpsR- and VpsT-dependent H-NS antirepression cascade.


Subject(s)
Bacterial Proteins/genetics , Biofilms/growth & development , Cyclic GMP/analogs & derivatives , DNA-Binding Proteins/genetics , Vibrio cholerae/physiology , Bacterial Proteins/metabolism , Cyclic GMP/metabolism , DNA-Binding Proteins/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation, Bacterial , Protein Binding , Protein Biosynthesis , Second Messenger Systems , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
16.
Biochem Biophys Res Commun ; 461(1): 65-9, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25849889

ABSTRACT

In Vibrio cholerae, the genes required for biofilm development are repressed by quorum sensing at high cell density due to the accumulation in the medium of two signaling molecules, cholera autoinducer 1 (CAI-1) and autoinducer 2 (AI-2). A significant fraction of toxigenic V. cholerae isolates, however, exhibit dysfunctional quorum sensing pathways. It was reported that transition state analogs of the enzyme methylthioadenosine/S-adenosylhomocysteine nucleosidase (MtnN) required to make AI-2 inhibited biofilm formation in the prototype quorum sensing-deficient strain N16961. This finding prompted us to examine the role of both autoinducers and MtnN in biofilm development and virulence gene expression in a quorum sensing-deficient genetic background. Here we show that deletion of mtnN encoding methylthioadenosine/S-adenosylhomocysteine nucleosidase, cqsA (CAI-1), and/or luxS (AI-2) do not prevent biofilm development. However, two independent mtnN mutants exhibited diminished growth rate and motility in swarm agar plates suggesting that, under certain conditions, MtnN could influence biofilm formation indirectly. Nevertheless, MtnN is not required for the development of a mature biofilm.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Carbon-Sulfur Lyases/metabolism , Ketones/metabolism , N-Glycosyl Hydrolases/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Quorum Sensing/physiology , Vibrio cholerae/physiology , Cell Movement/physiology
17.
PLoS One ; 10(2): e0118295, 2015.
Article in English | MEDLINE | ID: mdl-25679988

ABSTRACT

The histone-like nucleoid structuring protein (H-NS) functions as a transcriptional silencer by binding to AT-rich sequences at bacterial promoters. However, H-NS repression can be counteracted by other transcription factors in response to environmental changes. The identification of potential toxic factors, the expression of which is prevented by H-NS could facilitate the discovery of new regulatory proteins that may contribute to the emergence of new pathogenic variants by anti-silencing. Vibrio cholerae hns mutants of the El Tor biotype exhibit altered virulence, motility and environmental stress response phenotypes compared to wild type. We used an RNA-seq analysis approach to determine the basis of the above hns phenotypes and identify new targets of H-NS transcriptional silencing. H-NS affected the expression of 18% of all predicted genes in a growth phase-dependent manner. Loss of H-NS resulted in diminished expression of numerous genes encoding methyl-accepting chemotaxis proteins as well as chemotaxis toward the attractants glycine and serine. Deletion of hns also induced an endogenous envelope stress response resulting in elevated expression of rpoE encoding the extracytoplamic sigma factor E (σE). The RNA-seq analysis identified new genes directly repressed by H-NS that can affect virulence and biofilm development in the El Tor biotype cholera bacterium. We show that H-NS and the quorum sensing regulator HapR silence the transcription of the vieSAB three-component regulatory system in El Tor biotype V. cholerae. We also demonstrate that H-NS directly represses the transcription of hlyA (hemolysin), rtxCA (the repeat in toxin or RTX), rtxBDE (RTX transport) and the biosynthesis of indole. Of these genes, H-NS occupancy at the hlyA promoter was diminished by overexpression of the transcription activator HlyU. We discuss the role of H-NS transcriptional silencing in phenotypic differences exhibited by V. cholerae biotypes.


Subject(s)
Bacterial Proteins/genetics , Chemotaxis/genetics , Cholera/microbiology , DNA-Binding Proteins/genetics , Stress, Physiological/genetics , Vibrio cholerae/physiology , Vibrio cholerae/pathogenicity , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Bacterial , High-Throughput Nucleotide Sequencing , Indoles/metabolism , Mutation , Promoter Regions, Genetic , Protein Binding , Quorum Sensing/genetics , Transcription, Genetic , Virulence/genetics
18.
J Bacteriol ; 196(5): 1020-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24363348

ABSTRACT

Cholera is a waterborne diarrheal disease caused by Vibrio cholerae strains of serogroups O1 and O139. Expression of the general stress response regulator RpoS and formation of biofilm communities enhance the capacity of V. cholerae to persist in aquatic environments. The transition of V. cholerae between free-swimming (planktonic) and biofilm life-styles is regulated by the second messenger cyclic di-GMP (c-di-GMP). We previously reported that increasing the c-di-GMP pool by overexpression of a diguanylate cyclase diminished RpoS expression. Here we show that c-di-GMP repression of RpoS expression is eliminated by deletion of the genes vpsR and vpsT, encoding positive regulators of biofilm development. To determine the mechanism of this regulation, we constructed a strain expressing a vpsT-FLAG allele from native transcription and translation signals. Increasing the c-di-GMP pool induced vpsT-FLAG expression. The interaction between VpsT-FLAG and the rpoS promoter was demonstrated by chromatin immunoprecipitation. Furthermore, purified VpsT interacted with the rpoS promoter in a c-di-GMP-dependent manner. Primer extension analysis identified two rpoS transcription initiation sites located 43 bp (P1) and 63 bp (P2) upstream of the rpoS start codon. DNase I footprinting showed that the VpsT binding site at the rpoS promoter overlaps the primary P1 transcriptional start site. Deletion of vpsT significantly enhanced rpoS expression in V. cholerae biofilms that do not make HapR. This result suggests that VpsT and c-di-GMP contribute to the transcriptional silencing of rpoS in biofilms prior to cells entering the quorum-sensing mode.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Gene Expression Regulation, Bacterial/physiology , Sigma Factor/metabolism , Vibrio cholerae/metabolism , Vibrio cholerae/physiology , Bacterial Proteins/genetics , Cyclic GMP/analogs & derivatives , Cyclic GMP/genetics , Cyclic GMP/metabolism , Sigma Factor/genetics , Stress, Physiological/physiology , Transcription, Genetic/physiology , Vibrio cholerae/genetics
19.
Assay Drug Dev Technol ; 11(6): 382-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23906348

ABSTRACT

Quorum sensing is a cell-cell communication process in bacteria that involves the production, release, and subsequent detection of chemical signal molecules called autoinducers. In Vibrio cholerae, multiple input signals activate the expression of the quorum sensing regulator HapR, which acts to repress the expression of virulence factors. We have shown that CRP, the cyclic adenosine monophosphate (cAMP) receptor protein, enhances quorum sensing by activating the biosynthesis of cholera autoinducer 1, the major signaling molecule that contributes to the activation of HapR. Thus, proquorum sensing CRP agonists could inhibit virulence and lead to new drugs to treat severe cholera. In this study, we show that expression of the quorum sensing-regulated luxCDABE operon can be used as a robust readout for CRP activity. Further, we describe and validate a highly specific cell-based luminescence high-throughput screening assay for proquorum sensing CRP ligands. A pilot screen of 9,425 compounds yielded a hit rate of 0.02%, one hit being cAMP itself. The Z' value for this assay was 0.76 and its coefficient of variance 8% for the positive control compound. To our knowledge, this is the first cell-based assay for ligands of the highly conserved CRP protein of Gram-negative bacteria. The use of this assay to screen large chemical libraries could identify lead compounds to treat cholera, as well as small molecules to probe ligand-receptor interactions in the CRP molecule.


Subject(s)
Drug Evaluation, Preclinical/methods , Gram-Negative Bacteria/drug effects , High-Throughput Screening Assays/methods , Receptors, Cyclic AMP/agonists , Acyltransferases/genetics , Bacterial Proteins/genetics , Cholera/drug therapy , Cholera Toxin/antagonists & inhibitors , Drug Discovery , Ligands , Operon , Oxidoreductases/genetics , Quorum Sensing/drug effects
20.
Antimicrob Agents Chemother ; 57(8): 3950-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23733460

ABSTRACT

Vibrio cholerae strains of serogroups O1 and O139, the causative agents of the diarrheal illness cholera, express a single polar flagellum powered by sodium motive force and require motility to colonize and spread along the small intestine. In a previous study, we described a high-throughput assay for screening for small molecules that selectively inhibit bacterial motility and identified a family of quinazoline-2,4-diamino analogs (Q24DAs) that (i) paralyzed the sodium-driven polar flagellum of Vibrios and (ii) diminished cholera toxin secreted by El Tor biotype V. cholerae. In this study, we provide evidence that a Q24DA paralyzes the polar flagellum by interacting with the motor protein PomB. Inhibition of motility with the Q24DA enhanced the transcription of the cholera toxin genes in both biotypes. We also show that the Q24DA interacts with outer membrane protein OmpU and other porins to induce envelope stress and expression of the extracellular RNA polymerase sigma factor σ(E). We suggest that Q24DA-induced envelope stress could affect the correct folding, assembly, and secretion of pentameric cholera toxin in El Tor biotype V. cholerae independently of its effect on motility.


Subject(s)
Bacterial Proteins/metabolism , Flagella/drug effects , Quinazolines/pharmacology , Sodium Channels/metabolism , Stress, Physiological , Vibrio cholerae/drug effects , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Amino Acid Substitution , Bacterial Proteins/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholera Toxin/genetics , Cholera Toxin/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Flagella/physiology , Genes, Bacterial , Protein Folding , Protein Interaction Mapping , Sodium Channels/drug effects , Sodium Channels/genetics , Transcription, Genetic , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...