Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38853860

ABSTRACT

Attenuated strains of the intracellular pathogen Listeria monocytogenes can deliver genetically encoded payloads inside tumor cells. L. monocytogenes preferentially accumulates and propagates inside immune-suppressed tumor microenvironments. To maximize the payload impact in tumors and minimize damage to healthy tissues, it is desirable to induce payload synthesis when bacteria are eliminated from the healthy tissues but are grown to high numbers intratumorally. Here, we have engineered a tightly controlled gene expression system for intracellular L. monocytogenes inducible with a cumin derivative, cumate. Upon cumate addition, expression of a reporter gene is increased in L. monocytogenes growing in vitro by 80-fold, and in intracellular L. monocytogenes in murine tumors by 10-fold. This study demonstrates the feasibility of activating gene expression in intracellular bacteria in live animals using an edible inducer. The system is expected to enhance the efficacy and safety of the attenuated L. monocytogenes strains as antitumor payload delivery bacterial drones.

2.
Front Microbiol ; 14: 1258394, 2023.
Article in English | MEDLINE | ID: mdl-37928682

ABSTRACT

Major listeriosis outbreaks have been associated with fresh produce contaminated with Listeria monocytogenes. Strains that synthesize the Pss exopolysaccharide (EPS) have an estimated 102 to 104-fold advantage over nonsynthesizing strains in causing listeriosis. They more readily attach to the surfaces of fruit and vegetables forming EPS-biofilms that better withstand stresses associated with produce storage and consumption. Here, we show that the threat to fresh produce safety posed by the listerial EPS-biofilms may be countered by broadly available maple products. We serendipitously discovered that aqueous extracts of wood from several Acer (maple) and Carya (pecan, hickory) species inhibit the formation of listerial EPS-biofilms without affecting bacterial viability. One active ingredient in maple wood was identified as nortrachelogenin-8'-O-ß-D-glucopyranoside (NTG). At 120 µM, this lignan decreased colonization of the EPS-synthesizing L. monocytogenes on cantaloupe pieces by approximately 150-fold, and on cut celery and lettuce by 10 to 11-fold. Another lignan, lariciresinol, which is abundant in a common food sweetener, maple syrup, had antibiofilm activity comparable to that of NTG. Diluted in the range of 1:200 to 1:800 maple syrup from two random manufacturers prevented formation of listeiral EPS-biofilms. Importantly, not only did maple products drastically decrease colonization of fresh produce by the EPS-synthesizing strains, they also decreased, by 6 to 30-fold, colonization by the L. monocytogenes strains that do not synthesize measurable EPS, including strains from the infamous 2011 cantaloupe listeriosis outbreak. Inhibition of surface colonization by various listerial strains, broad availability of maple sap and syrup as well as maple lumber processing waste position maple products as potential antibiofilm agents for protecting fresh produce from L. monocytogenes.

3.
Front Microbiol ; 14: 1126940, 2023.
Article in English | MEDLINE | ID: mdl-37180237

ABSTRACT

Fresh produce contaminated with Listeria monocytogenes has caused major listeriosis outbreaks in the last decades. Our knowledge about components of the listerial biofilms formed on fresh produce and their roles in causing foodborne illness remains incomplete. Here, we investigated, for the first time, the role of the listerial Pss exopolysaccharide (EPS) in plant surface colonization and stress tolerance. Pss is the main component of L. monocytogenes biofilms synthesized at elevated levels of the second messenger c-di-GMP. We developed a new biofilm model, whereby L. monocytogenes EGD-e and its derivatives are grown in the liquid minimal medium in the presence of pieces of wood or fresh produce. After 48-h incubation, the numbers of colony forming units of the Pss-synthesizing strain on pieces of wood, cantaloupe, celery and mixed salads were 2-12-fold higher, compared to the wild-type strain. Colonization of manmade materials, metals and plastics, was largely unaffected by the presence of Pss. The biofilms formed by the EPS-synthesizing strain on cantaloupe rind were 6-16-fold more tolerant of desiccation, which resembles conditions of whole cantaloupe storage and transportation. Further, listeria in the EPS-biofilms survived exposure to low pH, a condition encountered by bacteria on the contaminated produce during passage through the stomach, by 11-116-fold better than the wild-type strain. We surmise that L. monocytogenes strains synthesizing Pss EPS have an enormous, 102-104-fold, advantage over the non-synthesizing strains in colonizing fresh produce, surviving during storage and reaching small intestines of consumers where they may cause disease. The magnitude of the EPS effect calls for better understanding of factors inducing Pss synthesis and suggests that prevention of listerial EPS-biofilms may significantly enhance fresh produce safety.

4.
Environ Microbiol ; 23(10): 5704-5715, 2021 10.
Article in English | MEDLINE | ID: mdl-34288318

ABSTRACT

Bacteria interact with fungi in a variety of ways to inhibit fungal growth, while the underlying mechanisms remain only partially characterized. The plant-beneficial Bacillus and Pseudomonas species are well-known antifungal biocontrol agents, whereas Lysobacter are far less studied. Members of Lysobacter are easy to grow in fermenters and are safe to humans, animals and plants. These environmentally ubiquitous bacteria use a diverse arsenal of weapons to prey on other microorganisms, including fungi and oomycetes. The small molecular toxins secreted by Lysobacter represent long-range weapons effective against filamentous fungi. The secreted hydrolytic enzymes act as intermediate-range weapons against non-filamentous fungi. The contact-dependent killing devices are proposed to work as short-range weapons. We describe here the structure, biosynthetic pathway, action mode and applications of one of the best-characterized long-range weapons, the heat-stable antifungal factor (HSAF) produced by Lysobacter enzymogenes. We discuss how the flagellar type III secretion system has evolved into an enzyme secretion machine for the intermediate-range antifungal weapons. We highlight an intricate mechanism coordinating the production of the long-range weapon, HSAF and the proposed contact-dependent killing device, type VI secretion system. We also overview the regulatory mechanisms of HSAF production involving specific transcription factors and the bacterial second messenger c-di-GMP.


Subject(s)
Lysobacter , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Bacterial Proteins/metabolism , Fungi/metabolism , Lysobacter/genetics , Lysobacter/metabolism , Transcription Factors/metabolism
5.
Mol Plant Pathol ; 22(5): 602-617, 2021 05.
Article in English | MEDLINE | ID: mdl-33709522

ABSTRACT

In the soil gammaproteobacterium Lysobacter enzymogenes, a natural fungal predator, the response regulator PilR controls type IV pili (T4P)-mediated twitching motility as well as synthesis of the heat-stable antifungal factor (HSAF). Earlier we showed that PilR acts via the second messenger, c-di-GMP; however, the mechanism remained unknown. Here, we describe how PilR, c-di-GMP signalling, and HSAF synthesis are connected. We screened genes for putative diguanylate cyclases (c-di-GMP synthases) and found that PilR binds to the promoter region of lchD and down-regulates its transcription. The DNA-binding affinity of PilR, and therefore its repressor function, are enhanced by phosphorylation by its cognate histidine kinase, PilS. The lchD gene product is a diguanylate cyclase, and the decrease in LchD levels shifts the ratio of c-di-GMP-bound and c-di-GMP-free transcription factor Clp, a key activator of the HSAF biosynthesis operon expression. Furthermore, Clp directly interacts with LchD and enhances its diguanylate cyclase activity. Therefore, the PilS-PilR two-component system activates T4P-motility while simultaneously decreasing c-di-GMP levels and promoting HSAF production via the highly specific LchD-c-di-GMP-Clp pathway. Coordinated increase in motility and secretion of the "long-distance" antifungal weapon HSAF is expected to ensure safer grazing of L. enzymogenes on soil or plant surfaces, unimpeded by fungal competitors, or to facilitate bacterial preying on killed fungal cells. This study uncovered the mechanism of coregulated pili-based motility and production of an antifungal antibiotic in L. enzymogenes, showcased the expanded range of functions of the PilS-PilR system, and highlighted exquisite specificity in c-di-GMP-mediated circuits.


Subject(s)
Antifungal Agents/metabolism , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/metabolism , Lysobacter/genetics , Phosphorus-Oxygen Lyases/metabolism , Transcription Factors/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Escherichia coli Proteins/genetics , Lysobacter/metabolism , Models, Biological , Phosphorus-Oxygen Lyases/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , Signal Transduction , Transcription Factors/genetics
6.
J Bacteriol ; 202(2)2020 01 02.
Article in English | MEDLINE | ID: mdl-31659012

ABSTRACT

Gene transfer agents (GTAs) are bacteriophage-like particles produced by several bacterial and archaeal lineages that contain small pieces of the producing cells' genomes that can be transferred to other cells in a process similar to transduction. One well-studied GTA is RcGTA, produced by the alphaproteobacterium Rhodobacter capsulatus RcGTA gene expression is regulated by several cellular regulatory systems, including the CckA-ChpT-CtrA phosphorelay. The transcription of multiple other regulator-encoding genes is affected by the response regulator CtrA, including genes encoding putative enzymes involved in the synthesis and hydrolysis of the second messenger bis-(3'-5')-cyclic dimeric GMP (c-di-GMP). To investigate whether c-di-GMP signaling plays a role in RcGTA production, we disrupted the CtrA-affected genes potentially involved in this process. We found that disruption of four of these genes affected RcGTA gene expression and production. We performed site-directed mutagenesis of key catalytic residues in the GGDEF and EAL domains responsible for diguanylate cyclase (DGC) and c-di-GMP phosphodiesterase (PDE) activities and analyzed the functions of the wild-type and mutant proteins. We also measured RcGTA production in R. capsulatus strains where intracellular levels of c-di-GMP were altered by the expression of either a heterologous DGC or a heterologous PDE. This adds c-di-GMP signaling to the collection of cellular regulatory systems controlling gene transfer in this bacterium. Furthermore, the heterologous gene expression and the four gene disruptions had similar effects on R. capsulatus flagellar motility as found for gene transfer, and we conclude that c-di-GMP inhibits both RcGTA production and flagellar motility in R. capsulatusIMPORTANCE Gene transfer agents (GTAs) are virus-like particles that move cellular DNA between cells. In the alphaproteobacterium Rhodobacter capsulatus, GTA production is affected by the activities of multiple cellular regulatory systems, to which we have now added signaling via the second messenger dinucleotide molecule bis-(3'-5')-cyclic dimeric GMP (c-di-GMP). Similar to the CtrA phosphorelay, c-di-GMP also affects R. capsulatus flagellar motility in addition to GTA production, with lower levels of intracellular c-di-GMP favoring increased flagellar motility and gene transfer. These findings further illustrate the interconnection of GTA production with global systems of regulation in R. capsulatus, providing additional support for the notion that the production of GTAs has been maintained in this and related bacteria because it provides a benefit to the producing organisms.


Subject(s)
Cyclic GMP/analogs & derivatives , Rhodobacter capsulatus/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclic GMP/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Gene Transfer, Horizontal/drug effects , Molecular Sequence Data , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/metabolism , Rhodobacter capsulatus/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Mol Plant Pathol ; 21(2): 218-229, 2020 02.
Article in English | MEDLINE | ID: mdl-31747123

ABSTRACT

YajQ, a binding protein of the universal bacterial second messenger cyclic di-GMP (c-di-GMP), affects virulence in several bacterial pathogens, including Xanthomonas campestris. In this bacterium, YajQ interacts with the transcription factor LysR. Upon c-di-GMP binding, the whole c-di-GMP-YajQ-LysR complex is found to dissociate from DNA, resulting in virulence gene regulation. Here, we identify a YajQ-LysR-like system in the bacterial biocontrol agent Lysobacter enzymogenes OH11 that secretes an antifungal antibiotic, heat-stable antifungal factor (HSAF) against crop fungal pathogens. We show that the YajQ homologue, CdgL (c-di-GMP receptor interacting with LysR) affects expression of the HSAF biosynthesis operon by interacting with the transcription activator LysR. The CdgL-LysR interaction enhances the apparent affinity of LysR to the promoter region upstream of the HSAF biosynthesis operon, which increases operon expression. Unlike the homologues CdgL (YajQ)-LysR system in X. campestris, we show that c-di-GMP binding to CdgL seems to weaken CdgL-LysR interactions and promote the release of CdgL from the LysR-DNA complex, which leads to decreased expression. Together, this study takes the YajQ-LysR-like system from bacterial pathogens to a crop-protecting bacterium that is able to regulate antifungal HSAF biosynthesis via disassembly of the c-di-GMP receptor-transcription activator complex.


Subject(s)
Antifungal Agents/metabolism , Cyclic GMP/analogs & derivatives , Lysobacter/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Xanthomonas campestris/metabolism
8.
Nat Chem Biol ; 15(9): 925-931, 2019 09.
Article in English | MEDLINE | ID: mdl-31406376

ABSTRACT

Multicellular organisms achieve greater complexity through cell divisions that generate different cell types. We engineered a simple genetic circuit that induces asymmetric cell division and subsequent cell differentiation in Escherichia coli. The circuit involves a scaffolding protein, PopZ, that is stably maintained at a single cell pole over multiple asymmetric cell divisions. PopZ was functionalized to degrade the signaling molecule, c-di-GMP. By regulating synthesis of functionalized PopZ via small molecules or light, we can chemically or optogenetically control the relative abundance of two distinct cell types, characterized by either low or high c-di-GMP levels. Differences in c-di-GMP levels can be transformed into genetically programmable differences in protein complex assembly or gene expression, which in turn produce differential behavior or biosynthetic activities. This study shows emergence of complex biological phenomena from a simple genetic circuit and adds programmable bacterial cell differentiation to the genetic toolbox of synthetic biology and biotechnology.


Subject(s)
Asymmetric Cell Division/physiology , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Bacterial Proteins/genetics , Cell Movement , Cloning, Molecular , DNA, Bacterial , Gene Expression Regulation, Bacterial , Signal Transduction
9.
ACS Synth Biol ; 8(6): 1314-1324, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31145854

ABSTRACT

Light in the near-infrared optical window (NIRW) penetrates deep through mammalian tissues, including the skull and brain tissue. Here we engineered an adenylate cyclase (AC) activated by NIRW light (NIRW-AC) and suitable for mammalian applications. To accomplish this goal, we constructed fusions of several bacteriophytochrome photosensory and bacterial AC modules using guidelines for designing chimeric homodimeric bacteriophytochromes. One engineered NIRW-AC, designated IlaM5, has significantly higher activity at 37 °C, is better expressed in mammalian cells, and can mediate cAMP-dependent photoactivation of gene expression in mammalian cells, in favorable contrast to the NIRW-ACs engineered earlier. The ilaM5 gene expressed from an AAV vector was delivered into the ventral basal thalamus region of the mouse brain, resulting in the light-controlled suppression of the cAMP-dependent wave pattern of the sleeping brain known as spindle oscillations. Reversible spindle oscillation suppression was observed in sleeping mice exposed to light from an external light source. This study confirms the robustness of principles of homodimeric bacteriophytochrome engineering, describes a NIRW-AC suitable for mammalian optogenetic applications, and demonstrates the feasibility of controlling brain activity via NIRW-ACs using transcranial irradiation.


Subject(s)
Adenylyl Cyclases , Infrared Rays , Optogenetics/methods , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/radiation effects , Animals , Brain/physiology , Cyclic AMP/metabolism , Electroencephalography , Mice , Neurons/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/radiation effects , Sleep/physiology
10.
J Bacteriol ; 200(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181123

ABSTRACT

Borrelia burgdorferi, the causative agent of Lyme disease, encounters two disparate host environments during its enzootic life cycle, Ixodes ticks and mammalian hosts. B. burgdorferi has a small genome that encodes a streamlined cyclic dimeric GMP (c-di-GMP) signaling system comprising a single diguanylate cyclase, Rrp1, and two phosphodiesterases. This system is essential for spirochete survival in ticks, in part because it controls the expression of the glp operon involved in glycerol utilization. In this study, we showed that a B. burgdorferi c-di-GMP receptor, PlzA, functions as both a positive and a negative regulator for glp expression. Deletion of plzA or mutation in plzA that impaired c-di-GMP binding abolished glp expression. On the other hand, overexpression of plzA resulted in glp repression, which could be rescued by simultaneous overexpression of rrp1. plzA overexpression in the rrp1 mutant, which is devoid of c-di-GMP, or overexpression of a plzA mutant incapable of c-di-GMP binding further enhanced glp repression. Combined results suggest that c-di-GMP-bound PlzA functions as a positive regulator, whereas ligand-free PlzA acts as a negative regulator for glp expression. Thus, PlzA of B. burgdorferi with a streamlined c-di-GMP signaling system not only controls multiple targets, as previously envisioned, but has also evolved different modes of action.IMPORTANCE The Lyme disease pathogen, Borrelia burgdorferi, has a simple cyclic dimeric GMP (c-di-GMP) signaling system essential for adaptation of the pathogen to the complicated tick environment. The c-di-GMP effector of B. burgdorferi, PlzA, has been shown to regulate multiple cellular processes, including motility, osmolality sensing, and nutrient utilization. The findings of this study demonstrate that PlzA not only controls multiple targets but also has different functional modalities, allowing it to act as both positive and negative regulator of the glp operon expression. This work highlights how bacteria with a small genome can compensate for the limited regulatory repertoire by increasing the complexity of targets and modes of action in their regulatory proteins.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Carrier Proteins/metabolism , Glycerol/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Bacterial , Intracellular Signaling Peptides and Proteins/genetics , Operon , Protein Binding , Signal Transduction
11.
Nucleic Acids Res ; 46(18): 9276-9288, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30202891

ABSTRACT

Enzymes controlling intracellular second messengers in bacteria, such as c-di-GMP, often affect some but not other targets. How such specificity is achieved is understood only partially. Here, we present a novel mechanism that enables specific c-di-GMP-dependent inhibition of the antifungal antibiotic production. Expression of the biosynthesis operon for Heat-Stable Antifungal Factor, HSAF, in Lysobacter enzymogenes occurs when the transcription activator Clp binds to two upstream sites. At high c-di-GMP levels, Clp binding to the lower-affinity site is compromised, which is sufficient to decrease gene expression. We identified a weak c-di-GMP phosphodiesterase, LchP, that plays a disproportionately high role in HSAF synthesis due to its ability to bind Clp. Further, Clp binding stimulates phosphodiesterase activity of LchP. An observation of a signaling complex formed by a c-di-GMP phosphodiesterase and a c-di-GMP-binding transcription factor lends support to the emerging paradigm that such signaling complexes are common in bacteria, and that bacteria and eukaryotes employ similar solutions to the specificity problem in second messenger-based signaling systems.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Cyclic GMP/analogs & derivatives , Lysobacter/metabolism , Signal Transduction , Antifungal Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Bacterial , Lysobacter/genetics , Models, Genetic , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Protein Binding , Protein Interaction Maps/genetics
12.
J Bacteriol ; 200(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29229701

ABSTRACT

Elevated levels of the second messenger c-di-GMP suppress virulence in diverse pathogenic bacteria, yet mechanisms are poorly characterized. In the foodborne pathogen Listeria monocytogenes, high c-di-GMP levels inhibit mammalian cell invasion. Here, we show that invasion is impaired because of the decreased expression levels of internalin genes whose products are involved in invasion. We further show that at high c-di-GMP levels, the expression of the entire virulence regulon is suppressed, and so is the expression of the prfA gene encoding the master activator of the virulence regulon. Analysis of mechanisms controlling prfA expression pointed to the transcription factor CodY as a c-di-GMP-sensitive component. In high-c-di-GMP strains, codY gene expression is decreased, apparently due to the lower activity of CodY, which functions as an activator of codY transcription. We found that listerial CodY does not bind c-di-GMP in vitro and therefore investigated whether c-di-GMP levels affect two known cofactors of listerial CodY, branched-chain amino acids and GTP. Our manipulation of branched-chain amino acid levels did not perturb the c-di-GMP effect; however, our replacement of listerial CodY with the streptococcal CodY homolog, whose activity is GTP independent, abolished the c-di-GMP effect. The results of this study suggest that elevated c-di-GMP levels decrease the activity of the coordinator of metabolism and virulence, CodY, possibly via lower GTP levels, and that decreased CodY activity suppresses L. monocytogenes virulence by the decreased expression of the PrfA virulence regulon.IMPORTANCEListeria monocytogenes is a pathogen causing listeriosis, a disease responsible for the highest mortality rate among foodborne diseases. Understanding how the virulence of this pathogen is regulated is important for developing treatments to decrease the frequency of listerial infections in susceptible populations. In this study, we describe the mechanism through which elevated levels of the second messenger c-di-GMP inhibit listerial invasion in mammalian cells. Inhibition is caused by the decreased activity of the transcription factor CodY that coordinates metabolism and virulence.


Subject(s)
Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Transcription Factors/genetics , Amino Acids, Branched-Chain/genetics , Amino Acids, Branched-Chain/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Cyclic GMP/analysis , Cyclic GMP/genetics , Cyclic GMP/metabolism , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , HT29 Cells , Host-Pathogen Interactions/genetics , Humans , Listeriosis/microbiology , Peptide Termination Factors/genetics , Promoter Regions, Genetic , Regulon , Virulence/genetics
13.
Methods Mol Biol ; 1657: 169-186, 2017.
Article in English | MEDLINE | ID: mdl-28889294

ABSTRACT

Signaling pathways involving second messenger c-di-GMP regulate various aspects of bacterial physiology and behavior. We describe the use of a red light-activated diguanylate cyclase (c-di-GMP synthase) and a blue light-activated c-di-GMP phosphodiesterase (hydrolase) for manipulating intracellular c-di-GMP levels in bacterial cells. We illustrate the application of these enzymes in regulating several c-di-GMP-dependent phenotypes, i.e., motility and biofilm phenotypes in E. coli and chemotactic behavior in the alphaproteobacterium Azospirillum brasilense. We expect these light-activated enzymes to be also useful in regulating c-di-GMP-dependent processes occurring at the fast timescale, for spatial control of bacterial populations, as well as for analyzing c-di-GMP-dependent phenomena at the single-cell level.


Subject(s)
Bacteria/metabolism , Bacteria/radiation effects , Bacterial Physiological Phenomena/radiation effects , Cyclic GMP/analogs & derivatives , Light , Bacteria/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms , Biomass , Chemotaxis , Cyclic GMP/metabolism , Enzyme Activators/radiation effects , Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/metabolism , Plasmids/genetics , Transformation, Bacterial
14.
Proteins ; 85(6): 1178, 2017 06.
Article in English | MEDLINE | ID: mdl-28504851
15.
Sci Transl Med ; 9(387)2017 04 26.
Article in English | MEDLINE | ID: mdl-28446687

ABSTRACT

A semiautonomous system enables implanted photoactivated cells to produce glucose-lowering hormones and maintain glucose homeostasis in diabetic mice (Shao et al, this issue).


Subject(s)
Glucose/metabolism , Animals , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide 1/pharmacology , Homeostasis/drug effects , Immunotherapy , Insulin/pharmacology , Mice
16.
J Bacteriol ; 199(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28264994

ABSTRACT

Bacterial chemotaxis receptors provide the sensory inputs that inform the direction of navigation in changing environments. Recently, we described the bacterial second messenger cyclic di-GMP (c-di-GMP) as a novel regulator of a subclass of chemotaxis receptors. In Azospirillum brasilense, c-di-GMP binds to a chemotaxis receptor, Tlp1, and modulates its signaling function during aerotaxis. Here, we further characterize the role of c-di-GMP in aerotaxis using a novel dichromatic optogenetic system engineered for manipulating intracellular c-di-GMP levels in real time. This system comprises a red/near-infrared-light-regulated diguanylate cyclase and a blue-light-regulated c-di-GMP phosphodiesterase. It allows the generation of transient changes in intracellular c-di-GMP concentrations within seconds of irradiation with appropriate light, which is compatible with the time scale of chemotaxis signaling. We provide experimental evidence that binding of c-di-GMP to the Tlp1 receptor activates its signaling function during aerotaxis, which supports the role of transient changes in c-di-GMP levels as a means of adjusting the response of A. brasilense to oxygen gradients. We also show that intracellular c-di-GMP levels in A. brasilense change with carbon metabolism. Our data support a model whereby c-di-GMP functions to imprint chemotaxis receptors with a record of recent metabolic experience, to adjust their contribution to the signaling output, thus allowing the cells to continually fine-tune chemotaxis sensory perception to their metabolic state.IMPORTANCE Motile bacteria use chemotaxis to change swimming direction in response to changes in environmental conditions. Chemotaxis receptors sense environmental signals and relay sensory information to the chemotaxis machinery, which ultimately controls the swimming pattern of cells. In bacteria studied to date, differential methylation has been known as a mechanism to control the activity of chemotaxis receptors and modulates their contribution to the overall chemotaxis response. Here, we used an optogenetic system to perturb intracellular concentrations of the bacterial second messenger c-di-GMP to show that in some chemotaxis receptors, c-di-GMP functions in a similar feedback loop to connect the metabolic status of the cells to the sensory activity of chemotaxis receptors.


Subject(s)
Azospirillum brasilense/physiology , Chemotaxis , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial , Locomotion , Carbon/metabolism , Cyclic GMP/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Light , Optogenetics/methods , Oxygen/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Phosphorus-Oxygen Lyases/genetics , Phosphorus-Oxygen Lyases/metabolism , Signal Transduction
17.
J Bacteriol ; 199(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28320886

ABSTRACT

Many aspects of bacterial physiology and behavior, including motility, surface attachment, and the cell cycle, are controlled by cyclic di-GMP (c-di-GMP)-dependent signaling pathways on the scale of seconds to minutes. Interrogation of such processes in real time requires tools for introducing rapid and reversible changes in intracellular c-di-GMP levels. Inducing the expression of genes encoding c-di-GMP-synthetic (diguanylate cyclases) and -degrading (c-di-GMP phosphodiesterase) enzymes by chemicals may not provide adequate temporal control. In contrast, light-controlled diguanylate cyclases and phosphodiesterases can be quickly activated and inactivated. A red/near-infrared-light-regulated diguanylate cyclase, BphS, was engineered previously, yet a complementary light-activated c-di-GMP phosphodiesterase has been lacking. In search of such a phosphodiesterase, we investigated two homologous proteins from Allochromatium vinosum and Magnetococcus marinus, designated BldP, which contain C-terminal EAL-BLUF modules, where EAL is a c-di-GMP phosphodiesterase domain and BLUF is a blue light sensory domain. Characterization of the BldP proteins in Escherichia coli and in vitro showed that they possess light-activated c-di-GMP phosphodiesterase activities. Interestingly, light activation in both enzymes was dependent on oxygen levels. The truncated EAL-BLUF fragment from A. vinosum BldP lacked phosphodiesterase activity, whereas a similar fragment from M. marinus BldP, designated EB1, possessed such activity that was highly (>30-fold) upregulated by light. Following light withdrawal, EB1 reverted to the inactive ground state with a half-life of ∼6 min. Therefore, the blue-light-activated phosphodiesterase EB1 can be used in combination with the red/near-infrared-light-regulated diguanylate cyclase BphS for the bidirectional regulation of c-di-GMP-dependent processes in E. coli as well as other bacterial and nonbacterial cells.IMPORTANCE Regulation of motility, attachment to surfaces, the cell cycle, and other bacterial processes controlled by the c-di-GMP signaling pathways occur at a fast (seconds-to-minutes) pace. Interrogation of these processes at high temporal and spatial resolution using chemicals is difficult or impossible, while optogenetic approaches may prove useful. We identified and characterized a robust, blue-light-activated c-di-GMP phosphodiesterase (hydrolase) that complements a previously engineered red/near-infrared-light-regulated diguanylate cyclase (c-di-GMP synthase). These two enzymes form a dichromatic module for manipulating intracellular c-di-GMP levels in bacterial and nonbacterial cells.


Subject(s)
Cyclic GMP/analogs & derivatives , Escherichia coli/metabolism , Escherichia coli/radiation effects , Genetics, Microbial/methods , Optogenetics/methods , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Alphaproteobacteria/enzymology , Alphaproteobacteria/genetics , Chromatiaceae/enzymology , Chromatiaceae/genetics , Cyclic GMP/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Light , Phosphoric Diester Hydrolases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Appl Environ Microbiol ; 83(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28087536

ABSTRACT

Lysobacter enzymogenes is a ubiquitous soil gammaproteobacterium that produces a broad-spectrum antifungal antibiotic, known as heat-stable antifungal factor (HSAF). To increase HSAF production for use against fungal crop diseases, it is important to understand how HSAF synthesis is regulated. To gain insights into transcriptional regulation of the HSAF synthesis gene cluster, we generated a library with deletion mutations in the genes predicted to encode response regulators of the two-component signaling systems in L. enzymogenes strain OH11. By quantifying HSAF production levels in the 45 constructed mutants, we identified two strains that produced significantly smaller amounts of HSAF. One of the mutations affected a gene encoding a conserved bacterial response regulator, PilR, which is commonly associated with type IV pilus synthesis. We determined that L. enzymogenes PilR regulates pilus synthesis and twitching motility via a traditional pathway, by binding to the pilA promoter and upregulating pilA expression. Regulation of HSAF production by PilR was found to be independent of pilus formation. We discovered that the pilR mutant contained significantly higher intracellular levels of the second messenger cyclic di-GMP (c-di-GMP) and that this was the inhibitory signal for HSAF production. Therefore, the type IV pilus regulator PilR in L. enzymogenes activates twitching motility while downregulating antibiotic HSAF production by increasing intracellular c-di-GMP levels. This study identifies a new role of a common pilus regulator in proteobacteria and provides guidance for increasing antifungal antibiotic production in L. enzymogenesIMPORTANCE PilR is a widespread response regulator of the two-component system known for regulating type IV pilus synthesis in proteobacteria. Here we report that, in the soil bacterium Lysobacter enzymogenes, PilR regulates pilus synthesis and twitching motility, as expected. Unexpectedly, PilR was also found to control intracellular levels of the second messenger c-di-GMP, which in turn inhibits production of the antifungal antibiotic HSAF. The coordinated production of type IV pili and antifungal antibiotics has not been observed previously.


Subject(s)
Antifungal Agents/metabolism , Cyclic GMP/analogs & derivatives , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Lysobacter/genetics , Lysobacter/metabolism , Soil Microbiology , Bacterial Proteins/genetics , Cyclic GMP/metabolism , Fimbriae, Bacterial/metabolism , Gene Library , Multigene Family , Mutation , Signal Transduction
19.
J Bacteriol ; 198(1): 7-11, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26148715

ABSTRACT

In recent years, Escherichia coli has served as one of a few model bacterial species for studying cyclic di-GMP (c-di-GMP) signaling. The widely used E. coli K-12 laboratory strains possess 29 genes encoding proteins with GGDEF and/or EAL domains, which include 12 diguanylate cyclases (DGC), 13 c-di-GMP-specific phosphodiesterases (PDE), and 4 "degenerate" enzymatically inactive proteins. In addition, six new GGDEF and EAL (GGDEF/EAL) domain-encoding genes, which encode two DGCs and four PDEs, have recently been found in genomic analyses of commensal and pathogenic E. coli strains. As a group of researchers who have been studying the molecular mechanisms and the genomic basis of c-di-GMP signaling in E. coli, we now propose a general and systematic dgc and pde nomenclature for the enzymatically active GGDEF/EAL domain-encoding genes of this model species. This nomenclature is intuitive and easy to memorize, and it can also be applied to additional genes and proteins that might be discovered in various strains of E. coli in future studies.


Subject(s)
Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/classification , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Terminology as Topic , Cyclic GMP/genetics , Cyclic GMP/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Signal Transduction
20.
Proteins ; 83(5): 799-804, 2015 May.
Article in English | MEDLINE | ID: mdl-25645367

ABSTRACT

The ability of bacteria to use cGMP as a second messenger has been controversial for decades. Recently, nucleotide cyclases from Rhodospirillum centenum, GcyA, and Xanthomonas campestris, GuaX, have been shown to possess guanylate cyclase activities. Enzymatic activities of these guanylate cyclases measured in vitro were low, which makes interpretation of the assays ambiguous. Protein sequence analysis at present is insufficient to distinguish between bacterial adenylate and guanylate cyclases, both of which belong to nucleotide cyclases of type III. We developed a simple method for discriminating between guanylate and adenylate cyclase activities in a physiologically relevant bacterial system. The method relies on the use of a mutant cAMP receptor protein, CRPG , constructed here. While wild-type CRP is activated exclusively by cAMP, CRPG can be activated by either cAMP or cGMP. Using CRP- and CRPG -dependent lacZ expression in two E. coli strains, we verified that R. centenum GcyA and X. campestris GuaX have primarily guanylate cyclase activities. Among two other bacterial nucleotide cyclases tested, one, GuaA from Azospillrillum sp. B510, proved to have guanylate cyclase activity, while the other one, Bradyrhizobium japonicum CyaA, turned out to function as an adenylate cyclase. The results obtained with this reporter system were in excellent agreement with direct measurements of cyclic nucleotides secreted by E. coli expressing nucleotide cyclase genes. The simple genetic screen developed here is expected to facilitate identification of bacterial guanylate cyclases and engineering of guanylate cyclases with desired properties.


Subject(s)
Bacterial Proteins/chemistry , Guanylate Cyclase/chemistry , Adenylyl Cyclases/chemistry , Cyclic GMP/chemistry , Enzyme Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...