Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.116
Filter
Add more filters

Publication year range
1.
Cell ; 178(6): 1299-1312.e29, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474368

ABSTRACT

Metformin is the first-line therapy for treating type 2 diabetes and a promising anti-aging drug. We set out to address the fundamental question of how gut microbes and nutrition, key regulators of host physiology, affect the effects of metformin. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we developed a high-throughput four-way screen to define the underlying host-microbe-drug-nutrient interactions. We show that microbes integrate cues from metformin and the diet through the phosphotransferase signaling pathway that converges on the transcriptional regulator Crp. A detailed experimental characterization of metformin effects downstream of Crp in combination with metabolic modeling of the microbiota in metformin-treated type 2 diabetic patients predicts the production of microbial agmatine, a regulator of metformin effects on host lipid metabolism and lifespan. Our high-throughput screening platform paves the way for identifying exploitable drug-nutrient-microbiome interactions to improve host health and longevity through targeted microbiome therapies. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Microbiome/drug effects , Host Microbial Interactions/drug effects , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Agmatine/metabolism , Animals , Caenorhabditis elegans/microbiology , Cyclic AMP Receptor Protein , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Longevity/drug effects , Metformin/pharmacology , Nutrients/metabolism
2.
Cell ; 169(7): 1168-1169, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28622501

ABSTRACT

The microbiome has emerged as a major determinant of the functioning of host organisms, affecting both health and disease. Here, Han et al. use the workhorse of aging research, C. elegans, to identify specific mechanisms by which gut bacteria influence mitochondrial dynamics and aging, a first step toward analogous manipulations to modulate human aging.


Subject(s)
Caenorhabditis elegans/microbiology , Longevity , Animals , Caenorhabditis elegans Proteins , Humans , Microbiota , Mitochondria
3.
Cell ; 169(7): 1249-1262.e13, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622510

ABSTRACT

Homeostasis of the gut microbiota critically influences host health and aging. Developing genetically engineered probiotics holds great promise as a new therapeutic paradigm to promote healthy aging. Here, through screening 3,983 Escherichia coli mutants, we discovered that 29 bacterial genes, when deleted, increase longevity in the host Caenorhabditis elegans. A dozen of these bacterial mutants also protect the host from age-related progression of tumor growth and amyloid-beta accumulation. Mechanistically, we discovered that five bacterial mutants promote longevity through increased secretion of the polysaccharide colanic acid (CA), which regulates mitochondrial dynamics and unfolded protein response (UPRmt) in the host. Purified CA polymers are sufficient to promote longevity via ATFS-1, the host UPRmt-responsive transcription factor. Furthermore, the mitochondrial changes and longevity effects induced by CA are conserved across different species. Together, our results identified molecular targets for developing pro-longevity microbes and a bacterial metabolite acting on host mitochondria to promote longevity.


Subject(s)
Caenorhabditis elegans/microbiology , Escherichia coli/genetics , Longevity , Aging/metabolism , Amyloid beta-Peptides/metabolism , Animals , Bacterial Load , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Escherichia coli/metabolism , Gene Deletion , Genome-Wide Association Study , Mitochondrial Dynamics , Models, Animal , Polysaccharides/metabolism , Transcription Factors/metabolism , Unfolded Protein Response
4.
Cell ; 169(3): 431-441.e8, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431244

ABSTRACT

The human microbiota greatly affects physiology and disease; however, the contribution of bacteria to the response to chemotherapeutic drugs remains poorly understood. Caenorhabditis elegans and its bacterial diet provide a powerful system to study host-bacteria interactions. Here, we use this system to study how bacteria affect the C. elegans response to chemotherapeutics. We find that different bacterial species can increase the response to one drug yet decrease the effect of another. We perform genetic screens in two bacterial species using three chemotherapeutic drugs: 5-fluorouracil (5-FU), 5-fluoro-2'-deoxyuridine (FUDR), and camptothecin (CPT). We find numerous bacterial nucleotide metabolism genes that affect drug efficacy in C. elegans. Surprisingly, we find that 5-FU and FUDR act through bacterial ribonucleotide metabolism to elicit their cytotoxic effects in C. elegans rather than by thymineless death or DNA damage. Our study provides a blueprint for characterizing the role of bacteria in the host response to chemotherapeutics.


Subject(s)
Antineoplastic Agents/metabolism , Caenorhabditis elegans/microbiology , Comamonas/metabolism , Escherichia coli/metabolism , Gastrointestinal Microbiome , Animals , Antineoplastic Agents/pharmacology , Camptothecin/metabolism , Camptothecin/pharmacology , Colorectal Neoplasms/drug therapy , Comamonas/genetics , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Deoxyuridine/pharmacology , Diet , Escherichia coli/genetics , Fluorouracil/metabolism , Fluorouracil/pharmacology , Humans , Models, Animal , Pyrimidine Nucleosides/metabolism
5.
Cell ; 159(2): 267-80, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303524

ABSTRACT

Discrimination between pathogenic and beneficial microbes is essential for host organism immunity and homeostasis. Here, we show that chemosensory detection of two secondary metabolites produced by Pseudomonas aeruginosa modulates a neuroendocrine signaling pathway that promotes avoidance behavior in the simple animal host Caenorhabditis elegans. Secondary metabolites phenazine-1-carboxamide and pyochelin activate a G-protein-signaling pathway in the ASJ chemosensory neuron pair that induces expression of the neuromodulator DAF-7/TGF-ß. DAF-7, in turn, activates a canonical TGF-ß signaling pathway in adjacent interneurons to modulate aerotaxis behavior and promote avoidance of pathogenic P. aeruginosa. Our data provide a chemical, genetic, and neuronal basis for how the behavior and physiology of a simple animal host can be modified by the microbial environment and suggest that secondary metabolites produced by microbes may provide environmental cues that contribute to pathogen recognition and host survival.


Subject(s)
Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Pseudomonas aeruginosa/metabolism , Animals , Behavior, Animal , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Neurons/metabolism , Neurosecretory Systems/physiology , Phenazines/metabolism , Phenols/metabolism , Species Specificity , Thiazoles/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
6.
Nature ; 613(7943): 324-331, 2023 01.
Article in English | MEDLINE | ID: mdl-36599989

ABSTRACT

Pathogens generate ubiquitous selective pressures and host-pathogen interactions alter social behaviours in many animals1-4. However, very little is known about the neuronal mechanisms underlying pathogen-induced changes in social behaviour. Here we show that in adult Caenorhabditis elegans hermaphrodites, exposure to a bacterial pathogen (Pseudomonas aeruginosa) modulates sensory responses to pheromones by inducing the expression of the chemoreceptor STR-44 to promote mating. Under standard conditions, C. elegans hermaphrodites avoid a mixture of ascaroside pheromones to facilitate dispersal5-13. We find that exposure to the pathogenic Pseudomonas bacteria enables pheromone responses in AWA sensory neurons, which mediate attractive chemotaxis, to suppress the avoidance. Pathogen exposure induces str-44 expression in AWA neurons, a process regulated by a transcription factor zip-5 that also displays a pathogen-induced increase in expression in AWA. STR-44 acts as a pheromone receptor and its function in AWA neurons is required for pathogen-induced AWA pheromone response and suppression of pheromone avoidance. Furthermore, we show that C. elegans hermaphrodites, which reproduce mainly through self-fertilization, increase the rate of mating with males after pathogen exposure and that this increase requires str-44 in AWA neurons. Thus, our results uncover a causal mechanism for pathogen-induced social behaviour plasticity, which can promote genetic diversity and facilitate adaptation of the host animals.


Subject(s)
Caenorhabditis elegans , Pheromones , Pseudomonas aeruginosa , Reproduction , Sexual Behavior, Animal , Animals , Female , Male , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Glycolipids/metabolism , Hermaphroditic Organisms/physiology , Pheromones/metabolism , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Receptors, Pheromone/metabolism , Reproduction/physiology , Sensory Receptor Cells/metabolism
7.
Cell ; 153(1): 228-39, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540700

ABSTRACT

The biguanide drug metformin is widely prescribed to treat type 2 diabetes and metabolic syndrome, but its mode of action remains uncertain. Metformin also increases lifespan in Caenorhabditis elegans cocultured with Escherichia coli. This bacterium exerts complex nutritional and pathogenic effects on its nematode predator/host that impact health and aging. We report that metformin increases lifespan by altering microbial folate and methionine metabolism. Alterations in metformin-induced longevity by mutation of worm methionine synthase (metr-1) and S-adenosylmethionine synthase (sams-1) imply metformin-induced methionine restriction in the host, consistent with action of this drug as a dietary restriction mimetic. Metformin increases or decreases worm lifespan, depending on E. coli strain metformin sensitivity and glucose concentration. In mammals, the intestinal microbiome influences host metabolism, including development of metabolic disease. Thus, metformin-induced alteration of microbial metabolism could contribute to therapeutic efficacy-and also to its side effects, which include folate deficiency and gastrointestinal upset.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Folic Acid/metabolism , Hypoglycemic Agents/pharmacology , Longevity/drug effects , Metformin/pharmacology , Methionine/metabolism , Adenylate Kinase/metabolism , Aging/drug effects , Animals , Biguanides/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caloric Restriction , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Escherichia coli/metabolism , Humans , Hypoglycemic Agents/metabolism , Metagenome , Metformin/metabolism , Transcription Factors/metabolism
8.
Immunity ; 48(5): 963-978.e3, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768179

ABSTRACT

Regulated antimicrobial peptide expression in the intestinal epithelium is key to defense against infection and to microbiota homeostasis. Understanding the mechanisms that regulate such expression is necessary for understanding immune homeostasis and inflammatory disease and for developing safe and effective therapies. We used Caenorhabditis elegans in a preclinical approach to discover mechanisms of antimicrobial gene expression control in the intestinal epithelium. We found an unexpected role for the cholinergic nervous system. Infection-induced acetylcholine release from neurons stimulated muscarinic signaling in the epithelium, driving downstream induction of Wnt expression in the same tissue. Wnt induction activated the epithelial canonical Wnt pathway, resulting in the expression of C-type lectin and lysozyme genes that enhanced host defense. Furthermore, the muscarinic and Wnt pathways are linked by conserved transcription factors. These results reveal a tight connection between the nervous system and the intestinal epithelium, with important implications for host defense, immune homeostasis, and cancer.


Subject(s)
Acetylcholine/immunology , Caenorhabditis elegans/immunology , Intestinal Mucosa/immunology , Wnt Signaling Pathway/immunology , Acetylcholine/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Bacteria/immunology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/metabolism , Gene Expression/immunology , Homeostasis/genetics , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/immunology , Neurons/metabolism , Wnt Signaling Pathway/genetics
9.
Cell ; 149(2): 452-66, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22500807

ABSTRACT

The nematode C. elegans is attracted to nutritious bacteria and is repelled by pathogens and toxins. Here we show that RNAi and toxin-mediated disruption of core cellular activities, including translation, respiration, and protein turnover, stimulate behavioral avoidance of normally attractive bacteria. RNAi of these and other essential processes induces expression of detoxification and innate immune effectors, even in the absence of toxins or pathogens. Disruption of core processes in non-neuronal tissues was sufficient to stimulate aversion behavior, revealing a neuroendocrine axis of control that additionally required serotonergic and Jnk kinase signaling pathways. We propose that surveillance pathways overseeing core cellular activities allow animals to detect invading pathogens that deploy toxins and virulence factors to undermine vital host functions. Variation in cellular surveillance and endocrine pathways controlling behavior, detoxification, and immunity selected by past toxin or microbial interactions could underlie aberrant responses to foods, medicines, and microbes.


Subject(s)
Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Host-Pathogen Interactions , Xenobiotics/metabolism , Animals , Bacteria/metabolism , Bacterial Toxins , Caenorhabditis elegans/cytology , Caenorhabditis elegans/immunology , Cell Physiological Phenomena , Immunity, Innate , MAP Kinase Signaling System , Neurosecretory Systems/metabolism , RNA Interference , Signal Transduction
10.
Nature ; 596(7871): 285-290, 2021 08.
Article in English | MEDLINE | ID: mdl-34321666

ABSTRACT

Ageing is driven by a loss of cellular integrity1. Given the major role of ubiquitin modifications in cell function2, here we assess the link between ubiquitination and ageing by quantifying whole-proteome ubiquitin signatures in Caenorhabditis elegans. We find a remodelling of the ubiquitinated proteome during ageing, which is ameliorated by longevity paradigms such as dietary restriction and reduced insulin signalling. Notably, ageing causes a global loss of ubiquitination that is triggered by increased deubiquitinase activity. Because ubiquitination can tag proteins for recognition by the proteasome3, a fundamental question is whether deficits in targeted degradation influence longevity. By integrating data from worms with a defective proteasome, we identify proteasomal targets that accumulate with age owing to decreased ubiquitination and subsequent degradation. Lowering the levels of age-dysregulated proteasome targets prolongs longevity, whereas preventing their degradation shortens lifespan. Among the proteasomal targets, we find the IFB-2 intermediate filament4 and the EPS-8 modulator of RAC signalling5. While increased levels of IFB-2 promote the loss of intestinal integrity and bacterial colonization, upregulation of EPS-8 hyperactivates RAC in muscle and neurons, and leads to alterations in the actin cytoskeleton and protein kinase JNK. In summary, age-related changes in targeted degradation of structural and regulatory proteins across tissues determine longevity.


Subject(s)
Aging/metabolism , Caenorhabditis elegans/metabolism , Proteome/metabolism , Ubiquitin/metabolism , Ubiquitination , Actin Cytoskeleton/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/metabolism , Cytoskeletal Proteins/metabolism , Intestines/microbiology , Longevity , Muscles/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proteome/chemistry , rac GTP-Binding Proteins/metabolism
11.
PLoS Genet ; 20(6): e1011324, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38875298

ABSTRACT

The Transforming Growth Factor beta (TGF-ß) family consists of numerous secreted peptide growth factors that play significant roles in cell function, tissue patterning, and organismal homeostasis, including wound repair and immunity. Typically studied as homodimers, these ligands have the potential to diversify their functions through ligand interactions that may enhance, repress, or generate novel functions. In the nematode Caenorhabditis elegans, there are only five TGF-ß ligands, providing an opportunity to dissect ligand interactions in fewer combinations than in vertebrates. As in vertebrates, these ligands can be divided into bone morphogenetic protein (BMP) and TGF-ß/Activin subfamilies that predominantly signal through discrete signaling pathways. The BMP subfamily ligand DBL-1 has been well studied for its role in the innate immune response in C. elegans. Here we show that all five TGF-ß ligands play a role in survival on bacterial pathogens. We also demonstrate that multiple TGF-ß ligand pairs act nonredundantly as part of this response. We show that the two BMP-like ligands-DBL-1 and TIG-2-function independently of each other in the immune response, while TIG-2/BMP and the TGF-ß/Activin-like ligand TIG-3 function together. Structural modeling supports the potential for TIG-2 and TIG-3 to form heterodimers. Additionally, we identify TIG-2 and TIG-3 as members of a rare subset of TGF-ß ligands lacking the conserved cysteine responsible for disulfide linking mature dimers. Finally, we show that canonical DBL-1/BMP receptor and Smad signal transducers function in the response to bacterial pathogens, while components of the DAF-7 TGF-ß/Activin signaling pathway do not play a major role in survival. These results demonstrate a novel potential for BMP and TGF-ß/Activin subfamily ligands to interact and may provide a mechanism for distinguishing the developmental and homeostatic functions of these ligands from an acute response such as the innate immune response to bacterial pathogens.


Subject(s)
Bone Morphogenetic Proteins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Immunity, Innate , Signal Transduction , Transforming Growth Factor beta , Animals , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/immunology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Immunity, Innate/genetics , Ligands , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Activins/metabolism , Activins/genetics , Neuropeptides
12.
Annu Rev Genet ; 52: 1-20, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30192637

ABSTRACT

My trajectory to becoming a plant biologist was shaped by a complex mix of scientific, political, sociological, and personal factors. I was trained as a microbiologist and molecular biologist in the late 1960s and early 1970s, a time of political upheaval surrounding the Vietnam War. My political activism taught me to be wary of the potential misuses of scientific knowledge and to promote the positive applications of science for the benefit of society. I chose agricultural science for my postdoctoral work. Because I was not trained as a plant biologist, I devised a postdoctoral project that took advantage of my microbiological training, and I explored using genetic technologies to transfer the ability to fix nitrogen from prokaryotic nitrogen-fixing species to the model plant Arabidopsis thaliana with the ultimate goal of engineering crop plants. The invention of recombinant DNA technology greatly facilitated the cloning and manipulation of bacterial nitrogen-fixation ( nif) genes, but it also forced me to consider how much genetic engineering of organisms, including human beings, is acceptable. My laboratory has additionally studied host-pathogen interactions using Arabidopsis and the nematode Caenorhabditis elegans as model hosts.


Subject(s)
Arabidopsis/genetics , Caenorhabditis elegans/genetics , Host-Pathogen Interactions/genetics , Symbiosis/genetics , Animals , Arabidopsis/microbiology , Biology/history , Caenorhabditis elegans/microbiology , History, 20th Century , History, 21st Century , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Nitrogen Fixation/genetics , Sinorhizobium meliloti/genetics , Sinorhizobium meliloti/pathogenicity
13.
Nature ; 584(7821): 410-414, 2020 08.
Article in English | MEDLINE | ID: mdl-32641833

ABSTRACT

In metazoans, the secreted proteome participates in intercellular signalling and innate immunity, and builds the extracellular matrix scaffold around cells. Compared with the relatively constant intracellular environment, conditions for proteins in the extracellular space are harsher, and low concentrations of ATP prevent the activity of intracellular components of the protein quality-control machinery. Until now, only a few bona fide extracellular chaperones and proteases have been shown to limit the aggregation of extracellular proteins1-5. Here we performed a systematic analysis of the extracellular proteostasis network in Caenorhabditis elegans with an RNA interference screen that targets genes that encode the secreted proteome. We discovered 57 regulators of extracellular protein aggregation, including several proteins related to innate immunity. Because intracellular proteostasis is upregulated in response to pathogens6-9, we investigated whether pathogens also stimulate extracellular proteostasis. Using a pore-forming toxin to mimic a pathogenic attack, we found that C. elegans responded by increasing the expression of components of extracellular proteostasis and by limiting aggregation of extracellular proteins. The activation of extracellular proteostasis was dependent on stress-activated MAP kinase signalling. Notably, the overexpression of components of extracellular proteostasis delayed ageing and rendered worms resistant to intoxication. We propose that enhanced extracellular proteostasis contributes to systemic host defence by maintaining a functional secreted proteome and avoiding proteotoxicity.


Subject(s)
Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Extracellular Space/metabolism , Protein Aggregates , Proteostasis , Aging/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , MAP Kinase Signaling System , Protein Aggregation, Pathological/prevention & control , Proteome/genetics , Proteome/metabolism , RNA Interference
14.
Nature ; 586(7829): 445-451, 2020 10.
Article in English | MEDLINE | ID: mdl-32908307

ABSTRACT

Caenorhabditis elegans must distinguish pathogens from nutritious food sources among the many bacteria to which it is exposed in its environment1. Here we show that a single exposure to purified small RNAs isolated from pathogenic Pseudomonas aeruginosa (PA14) is sufficient to induce pathogen avoidance in the treated worms and in four subsequent generations of progeny. The RNA interference (RNAi) and PIWI-interacting RNA (piRNA) pathways, the germline and the ASI neuron are all required for avoidance behaviour induced by bacterial small RNAs, and for the transgenerational inheritance of this behaviour. A single P. aeruginosa non-coding RNA, P11, is both necessary and sufficient to convey learned avoidance of PA14, and its C. elegans target, maco-1, is required for avoidance. Our results suggest that this non-coding-RNA-dependent mechanism evolved to survey the microbial environment of the worm, use this information to make appropriate behavioural decisions and pass this information on to its progeny.


Subject(s)
Avoidance Learning , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Pseudomonas aeruginosa/genetics , RNA, Bacterial/genetics , RNA, Untranslated/genetics , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neurons/metabolism , Pseudomonas aeruginosa/pathogenicity , RNA Interference , RNA, Small Interfering/genetics , Ribonuclease III/metabolism , Species Specificity , Transforming Growth Factor beta/metabolism , Wills
15.
Nature ; 583(7816): 415-420, 2020 07.
Article in English | MEDLINE | ID: mdl-32555456

ABSTRACT

Animals coexist in commensal, pathogenic or mutualistic relationships with complex communities of diverse organisms, including microorganisms1. Some bacteria produce bioactive neurotransmitters that have previously been proposed to modulate nervous system activity and behaviours of their hosts2,3. However, the mechanistic basis of this microbiota-brain signalling and its physiological relevance are largely unknown. Here we show that in Caenorhabditis elegans, the neuromodulator tyramine produced by commensal Providencia bacteria, which colonize the gut, bypasses the requirement for host tyramine biosynthesis and manipulates a host sensory decision. Bacterially produced tyramine is probably converted to octopamine by the host tyramine ß-hydroxylase enzyme. Octopamine, in turn, targets the OCTR-1 octopamine receptor on ASH nociceptive neurons to modulate an aversive olfactory response. We identify the genes that are required for tyramine biosynthesis in Providencia, and show that these genes are necessary for the modulation of host behaviour. We further find that C. elegans colonized by Providencia preferentially select these bacteria in food choice assays, and that this selection bias requires bacterially produced tyramine and host octopamine signalling. Our results demonstrate that a neurotransmitter produced by gut bacteria mimics the functions of the cognate host molecule to override host control of a sensory decision, and thereby promotes fitness of both the host and the microorganism.


Subject(s)
Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Feeding Behavior/physiology , Intestines/microbiology , Neurotransmitter Agents/metabolism , Providencia/metabolism , Smell/physiology , Animals , Avoidance Learning/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gastrointestinal Microbiome/physiology , Metabolomics , Mutation , Octanols/pharmacology , Octopamine/biosynthesis , Octopamine/metabolism , Providencia/enzymology , Providencia/physiology , Receptors, Biogenic Amine/metabolism , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/metabolism , Smell/drug effects , Tyramine/biosynthesis , Tyramine/metabolism , Tyrosine Decarboxylase/deficiency , Tyrosine Decarboxylase/genetics
16.
PLoS Pathog ; 19(2): e1010925, 2023 02.
Article in English | MEDLINE | ID: mdl-36800381

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa PAO1 is infected by the filamentous bacteriophage Pf4. Pf4 virions promote biofilm formation, protect bacteria from antibiotics, and modulate animal immune responses in ways that promote infection. Furthermore, strains cured of their Pf4 infection (ΔPf4) are less virulent in animal models of infection. Consistently, we find that strain ΔPf4 is less virulent in a Caenorhabditis elegans nematode infection model. However, our data indicate that PQS quorum sensing is activated and production of the pigment pyocyanin, a potent virulence factor, is enhanced in strain ΔPf4. The reduced virulence of ΔPf4 despite high levels of pyocyanin production may be explained by our finding that C. elegans mutants unable to sense bacterial pigments through the aryl hydrocarbon receptor are more susceptible to ΔPf4 infection compared to wild-type C. elegans. Collectively, our data support a model where suppression of quorum-regulated virulence factors by Pf4 allows P. aeruginosa to evade detection by innate host immune responses.


Subject(s)
Inovirus , Pseudomonas Phages , Animals , Pseudomonas aeruginosa , Caenorhabditis elegans/microbiology , Pyocyanine , Quorum Sensing , Virulence Factors , Biofilms , Anti-Bacterial Agents/pharmacology , Bacterial Proteins
17.
PLoS Pathog ; 19(10): e1011730, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37906605

ABSTRACT

Sphingolipids are required for diverse biological functions and are degraded by specific catabolic enzymes. However, the mechanisms that regulate sphingolipid catabolism are not known. Here we characterize a transcriptional axis that regulates sphingolipid breakdown to control resistance against bacterial infection. From an RNAi screen for transcriptional regulators of pathogen resistance in the nematode C. elegans, we identified the nuclear hormone receptor nhr-66, a ligand-gated transcription factor homologous to human hepatocyte nuclear factor 4. Tandem chromatin immunoprecipitation-sequencing and RNA sequencing experiments revealed that NHR-66 is a transcriptional repressor, which directly targets sphingolipid catabolism genes. Transcriptional de-repression of two sphingolipid catabolic enzymes in nhr-66 loss-of-function mutants drives the breakdown of sphingolipids, which enhances host susceptibility to infection with the bacterial pathogen Pseudomonas aeruginosa. These data define transcriptional control of sphingolipid catabolism in the regulation of cellular sphingolipids, a process that is necessary for pathogen resistance.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Humans , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Transcription Factors/metabolism , Gene Expression Regulation , Sphingolipids/genetics , Sphingolipids/metabolism
18.
PLoS Pathog ; 19(7): e1011507, 2023 07.
Article in English | MEDLINE | ID: mdl-37440595

ABSTRACT

Pore-forming toxins (PFTs) are effective tools for pathogens infection. By disrupting epithelial barriers and killing immune cells, PFTs promotes the colonization and reproduction of pathogenic microorganisms in their host. In turn, the host triggers defense responses, such as endocytosis, exocytosis, or autophagy. Bacillus thuringiensis (Bt) bacteria produce PFT, known as crystal proteins (Cry) which damage the intestinal cells of insects or nematodes, eventually killing them. In insects, aminopeptidase N (APN) has been shown to act as an important receptor for Cry toxins. Here, using the nematode Caenorhabditis elegans as model, an extensive screening of APN gene family was performed to analyze the potential role of these proteins in the mode of action of Cry5Ba against the nematode. We found that one APN, MNP-1, participate in the toxin defense response, since the mnp-1(ok2434) mutant showed a Cry5Ba hypersensitive phenotype. Gene expression analysis in mnp-1(ok2434) mutant revealed the involvement of two protease genes, F19C6.4 and R03G8.6, that participate in Cry5Ba degradation. Finally, analysis of the transduction pathway involved in F19C6.4 and R03G8.6 expression revealed that upon Cry5Ba exposure, the worms up regulated both protease genes through the activation of the FOXO transcription factor DAF-16, which was translocated into the nucleus. The nuclear location of DAF-16 was found to be dependent on mnp-1 under Cry5Ba treatment. Our work provides evidence of new host responses against PFTs produced by an enteric pathogenic bacterium, resulting in activation of host intestinal proteases that degrade the PFT in the intestine.


Subject(s)
Bacillus thuringiensis , Caenorhabditis elegans Proteins , Animals , Caenorhabditis elegans/microbiology , Peptide Hydrolases/metabolism , Aminopeptidases/metabolism , Endotoxins/metabolism , Caenorhabditis elegans Proteins/metabolism , Hemolysin Proteins/metabolism , Intestines , Endopeptidases/metabolism , Bacterial Proteins/metabolism , Bacillus thuringiensis/metabolism , Forkhead Transcription Factors/metabolism
19.
PLoS Genet ; 18(3): e1010103, 2022 03.
Article in English | MEDLINE | ID: mdl-35275914

ABSTRACT

Monitoring mitochondrial function is crucial for organismal survival. This task is performed by mitochondrial surveillance or quality control pathways, which are activated by signals originating from mitochondria and relayed to the nucleus (retrograde response) to start transcription of protective genes. In Caenorhabditis elegans, several systems are known to play this role, including the UPRmt, MAPKmt, and the ESRE pathways. These pathways are highly conserved and their loss compromises survival following mitochondrial stress. In this study, we found a novel interaction between the box C/D snoRNA core proteins (snoRNPs) and mitochondrial surveillance and innate immune pathways. We showed that box C/D, but not box H/ACA, snoRNPs are required for the full function of UPRmt and ESRE upon stress. The loss of box C/D snoRNPs reduced mitochondrial mass, mitochondrial membrane potential, and oxygen consumption rate, indicating overall degradation of mitochondrial function. Concomitantly, the loss of C/D snoRNPs increased immune response and reduced host intestinal colonization by infectious bacteria, improving host resistance to pathogenesis. Our data may indicate a model wherein box C/D snoRNP machinery regulates a "switch" of the cell's activity between mitochondrial surveillance and innate immune activation. Understanding this mechanism is likely to be important for understanding multifactorial processes, including responses to infection and aging.


Subject(s)
Mitochondria , Ribonucleoproteins, Small Nucleolar , Animals , Caenorhabditis elegans/microbiology , Immunity, Innate/genetics , Mitochondria/genetics , Mitochondria/metabolism , RNA, Small Nucleolar , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism
20.
J Bacteriol ; 206(5): e0040223, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38687034

ABSTRACT

Pseudomonas aeruginosa is an opportunistic bacterial pathogen that commonly causes medical hardware, wound, and respiratory infections. Temperate filamentous Pf phages that infect P. aeruginosa impact numerous virulence phenotypes. Most work on Pf phages has focused on Pf4 and its host P. aeruginosa PAO1. Expanding from Pf4 and PAO1, this study explores diverse Pf phages infecting P. aeruginosa clinical isolates. We describe a simple technique targeting the Pf lysogeny maintenance gene, pflM (PA0718), that enables the effective elimination of Pf prophages from diverse P. aeruginosa hosts. The pflM gene shows diversity among different Pf phage isolates; however, all examined pflM alleles encode the DUF5447 domain. We demonstrate that pflM deletion results in prophage excision but not replication, leading to total prophage loss, indicating a role for lysis/lysogeny decisions for the DUF5447 domain. This study also assesses the effects different Pf phages have on host quorum sensing, biofilm formation, pigment production, and virulence against the bacterivorous nematode Caenorhabditis elegans. We find that Pf phages have strain-specific impacts on quorum sensing and biofilm formation, but nearly all suppress pigment production and increase C. elegans avoidance behavior. Collectively, this research not only introduces a valuable tool for Pf prophage elimination from diverse P. aeruginosa isolates but also advances our understanding of the complex relationship between P. aeruginosa and filamentous Pf phages.IMPORTANCEPseudomonas aeruginosa is an opportunistic bacterial pathogen that is frequently infected by filamentous Pf phages (viruses) that integrate into its chromosome, affecting behavior. Although prior work has focused on Pf4 and PAO1, this study investigates diverse Pf in clinical isolates. A simple method targeting the deletion of the Pf lysogeny maintenance gene pflM (PA0718) effectively eliminates Pf prophages from clinical isolates. The research evaluates the impact Pf prophages have on bacterial quorum sensing, biofilm formation, and virulence phenotypes. This work introduces a valuable tool to eliminate Pf prophages from clinical isolates and advances our understanding of P. aeruginosa and filamentous Pf phage interactions.


Subject(s)
Prophages , Pseudomonas aeruginosa , Quorum Sensing , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Prophages/genetics , Prophages/physiology , Virulence , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/virology , Biofilms/growth & development , Animals , Lysogeny , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL