1
|
Kim D, Trang K, Pees B, Karimzadegan S, Bodkhe R, Hammond S, Shapira M. Identification of intestinal mediators of Caenorhabditis elegans DBL-1/BMP immune signaling shaping gut microbiome composition. mBio 2025:e0370324. [PMID: 39878514 DOI: 10.1128/mbio.03703-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
The composition of the gut microbiome is determined by a complex interplay of diet, host genetics, microbe-microbe interactions, abiotic factors, and stochasticity. Previous studies have demonstrated the importance of host genetics in community assembly of the Caenorhabditis elegans gut microbiome and identified a central role for DBL-1/BMP immune signaling in determining the abundance of gut Enterobacteriaceae. However, the effects of DBL-1 signaling on gut bacteria were found to depend on its activation in extra-intestinal tissues, highlighting a gap in our understanding of the proximal factors that determine microbiome composition. In the present study, we used RNA-seq gene expression analysis of wildtype, dbl-1 and sma-3 mutants, and dbl-1 over-expressors to identify candidate DBL-1/BMP targets that may mediate the pathway's effects on gut commensals. Bacterial colonization experiments in mutants, or following RNAi-mediated knock-down of candidate genes specifically in the intestine, demonstrated their local contribution to intestinal control of Enterobacteriaceae abundance. Furthermore, epistasis analysis suggested that these contributions were downstream of the DBL-1 pathway, together suggesting that examined candidates were intestinal effectors and mediators of DBL-1 signaling, contributing to the shaping of gut microbiome composition.IMPORTANCECompared to the roles of diet, environmental availability, or lifestyle in determining gut microbiome composition, that of genetic factors is the least understood and often underestimated. The identification of intestinal effectors of distinct molecular functions that control enteric bacteria offers a glimpse into the genetic logic of microbiome control as well as a list of targets for future exploration of this logic.
Collapse
Affiliation(s)
- Dan Kim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Kenneth Trang
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Barbara Pees
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Siavash Karimzadegan
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Rahul Bodkhe
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Sabrina Hammond
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| | - Michael Shapira
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
2
|
Peedikayil-Kurien S, Haque R, Gat A, Oren-Suissa M. Modulation by NPY/NPF-like receptor underlies experience-dependent, sexually dimorphic learning. Nat Commun 2025; 16:662. [PMID: 39809755 PMCID: PMC11733012 DOI: 10.1038/s41467-025-55950-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
The evolutionary paths taken by each sex within a given species sometimes diverge, resulting in behavioral differences. Given their distinct needs, the mechanism by which each sex learns from a shared experience is still an open question. Here, we reveal sexual dimorphism in learning: C. elegans males do not learn to avoid the pathogenic bacteria PA14 as efficiently and rapidly as hermaphrodites. Notably, neuronal activity following pathogen exposure was dimorphic: hermaphrodites generate robust representations, while males, in line with their behavior, exhibit contrasting representations. Transcriptomic and behavioral analysis revealed that the neuropeptide receptor npr-5, an ortholog of the mammalian NPY/NPF-like receptor, regulates male learning by modulating neuronal activity. Furthermore, we show the dependency of the males' decision-making on their sexual status and demonstrate the role of npr-5 as a modulator of incoming sensory cues. Taken together, these findings illustrate how neuromodulators drive sex-specific behavioral plasticity in response to a shared experience.
Collapse
Affiliation(s)
- Sonu Peedikayil-Kurien
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Rizwanul Haque
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Asaf Gat
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Meital Oren-Suissa
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel.
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
3
|
Schaller ML, Sykes MM, Easow SA, Carranza FR, Tuckowski AM, Shah YM, Leiser SF. Perception of Enterococcus faecalis without infection induces fmo-2 in C. elegans. MICROPUBLICATION BIOLOGY 2025; 2025. [PMID: 39867229 PMCID: PMC11759934 DOI: 10.17912/micropub.biology.001422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025]
Abstract
C. elegans pathogenic susceptibility is influenced by the worm's detection of its environment and its capacity to resist and resolve damage following infection. Here, we use a model where worms can sense, but not ingest, the pathogen Enterococcus faecalis (EF) . We identify that perception of EF without infection induces the stress-response gene fmo-2. We further identify that neural and intestinal signaling genes are necessary for fmo-2 induction without active infection. Finally, we show that fmo-2 overexpression is sufficient to extend lifespan with EF exposure, while fmo-2 KO is not detrimental, suggesting that additional fmo-2 expression benefits worms in this condition.
Collapse
Affiliation(s)
- Megan L Schaller
- Molecular and Integrative Physiology Department, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Madeline M Sykes
- Department of Molecular and Cellular Pathology, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Sarah A Easow
- Molecular and Integrative Physiology Department, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Faith R Carranza
- Cellular and Molecular Biology Program, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Angela M Tuckowski
- Cellular and Molecular Biology Program, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Yatrik M Shah
- Molecular and Integrative Physiology Department, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| | - Scott F Leiser
- Molecular and Integrative Physiology Department, University of Michigan-Ann Arbor, Ann Arbor, Michigan, United States
| |
Collapse
|
4
|
Biswas K, Moore C, Rogers H, Wani KA, Pukkila-Worley R, Higgins DP, Walker AK, Rand JB, Francis MM. Transcriptional responses to prolonged oxidative stress require cholinergic activation of G-protein-coupled receptor signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.628021. [PMID: 39829818 PMCID: PMC11741395 DOI: 10.1101/2025.01.06.628021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Organisms have evolved protective strategies that are geared toward limiting cellular damage and enhancing organismal survival in the face of environmental stresses, but how these protective mechanisms are coordinated remains unclear. Here, we define a requirement for neural activity in mobilizing the antioxidant defenses of the nematode Caenorhabditis elegans both during prolonged oxidative stress and prior to its onset. We show that acetylcholine-deficient mutants are particularly vulnerable to prolonged oxidative stress. We find that prolonged oxidative stress mobilizes a broad transcriptional response which is strongly dependent on both cholinergic signaling and activation of the muscarinic G-protein acetylcholine coupled receptor (mAChR) GAR-3. Gene enrichment analysis revealed a lack of upregulation of proteasomal proteolysis machinery in both cholinergic-deficient and gar-3 mAChR mutants, suggesting that muscarinic activation is critical for stress-responsive upregulation of protein degradation pathways. Further, we find that GAR-3 overexpression in cholinergic motor neurons prolongs survival during prolonged oxidative stress. Our studies demonstrate neuronal modulation of antioxidant defenses through cholinergic activation of G protein-coupled receptor signaling pathways, defining new potential links between cholinergic signaling, oxidative damage, and neurodegenerative disease.
Collapse
|
5
|
Will I, Stevens EJ, Belcher T, King KC. 'Re-Wilding' an Animal Model With Microbiota Shifts Immunity and Stress Gene Expression During Infection. Mol Ecol 2025; 34:e17586. [PMID: 39529601 DOI: 10.1111/mec.17586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The frequency of emerging disease is growing with ongoing human activity facilitating new host-pathogen interactions. Novel infection outcomes can also be shaped by the host microbiota. Caenorhabditis elegans nematodes experimentally colonised by a wild microbiota community and infected by the widespread animal pathogen, Staphylococcus aureus, have been shown to suffer higher mortality than those infected by the pathogen alone. Understanding the host responses to such microbiota-pathogen ecological interactions is key to pinpointing the mechanism underlying severe infection outcomes. We conducted transcriptomic analyses of C. elegans colonised by its native microbiota, S. aureus and both in combination. Correlations between altered collagen gene expression and heightened mortality in co-colonised hosts suggest the microbiota modified host resistance to infection. Furthermore, microbiota colonised hosts showed increased expression of immunity genes and variable expression of stress response genes during infection. Changes in host immunity and stress response could encompass both causes and effects of severe infection outcomes. 'Re-wilding' this model nematode host with its native microbiota indicated that typically commensal microbes can mediate molecular changes in the host that are costly when challenged by a novel emerging pathogen.
Collapse
Affiliation(s)
- Ian Will
- Department of Biology, University of Oxford, Oxford, UK
| | - Emily J Stevens
- Department of Biology, University of Oxford, Oxford, UK
- School of Life Sciences, Keele University, Newcastle-under-Lyme, UK
| | | | - Kayla C King
- Department of Biology, University of Oxford, Oxford, UK
- Department of Zoology, University of British Columbia, Vancouver, Canada
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Liu K, Grover M, Trusch F, Vagena-Pantoula C, Ippolito D, Barkoulas M. Paired C-type lectin receptors mediate specific recognition of divergent oomycete pathogens in C. elegans. Cell Rep 2024; 43:114906. [PMID: 39460939 DOI: 10.1016/j.celrep.2024.114906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/16/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Innate immune responses can be triggered upon detection of pathogen- or damage-associated molecular patterns by host receptors that are often present on the surface of immune cells. While invertebrates like Caenorhabditis elegans lack professional immune cells, they still mount pathogen-specific responses. However, the identity of host receptors in the nematode remains poorly understood. Here, we show that C-type lectin receptors mediate species-specific recognition of divergent oomycetes in C. elegans. A CLEC-27/CLEC-35 pair is essential for recognition of the oomycete Myzocytiopsis humicola, while a CLEC-26/CLEC-36 pair is required for detection of Haptoglossa zoospora. Both clec pairs are transcriptionally regulated through a shared promoter by the conserved PRD-like homeodomain transcription factor CEH-37/OTX2 and act in sensory neurons and the anterior intestine to trigger a protective immune response in the epidermis. This system enables redundant tissue sensing of oomycete threats through canonical CLEC receptors and host defense via cross-tissue communication.
Collapse
Affiliation(s)
- Kenneth Liu
- Department of Life Sciences, Imperial College, SW7 2AZ London, UK
| | - Manish Grover
- Department of Life Sciences, Imperial College, SW7 2AZ London, UK
| | - Franziska Trusch
- Department of Life Sciences, Imperial College, SW7 2AZ London, UK
| | | | | | | |
Collapse
|
7
|
Wang Y, Sun X, Feng L, Zhang K, Yang W. Nervous system guides behavioral immunity in Caenorhabditis elegans. PeerJ 2024; 12:e18289. [PMID: 39430568 PMCID: PMC11488496 DOI: 10.7717/peerj.18289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
Caenorhabditis elegans is a versatile model organism for exploring complex biological systems. Microbes and the external environment can affect the nervous system and drive behavioral changes in C. elegans. For better survival, C. elegans may develop behavioral immunity to avoid potential environmental pathogens. However, the molecular and cellular mechanisms underlying this avoidance behavior are not fully understood. The dissection of sensorimotor circuits in behavioral immunity may promote advancements in research on the neuronal connectome in uncovering neuronal regulators of behavioral immunity. In this review, we discuss how the nervous system coordinates behavioral immunity by translating various pathogen-derived cues and physiological damage to motor output in response to pathogenic threats in C. elegans. This understanding may provide insights into the fundamental principles of immune strategies that can be applied across species and potentially contribute to the development of novel therapies for immune-related diseases.
Collapse
Affiliation(s)
- Yu Wang
- Department of Physiology/West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xuehong Sun
- Department of Forensic Pathology/West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Lixiang Feng
- Department of Physiology/West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Kui Zhang
- Department of Forensic Pathology/West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Wenxing Yang
- Department of Physiology/West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Castiglioni VG, Olmo-Uceda MJ, Villena-Giménez A, Muñoz-Sánchez JC, Legarda EG, Elena SF. Story of an infection: Viral dynamics and host responses in the Caenorhabditis elegans-Orsay virus pathosystem. SCIENCE ADVANCES 2024; 10:eadn5945. [PMID: 39331715 PMCID: PMC11430451 DOI: 10.1126/sciadv.adn5945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
Orsay virus (OrV) is the only known natural virus affecting Caenorhabditis elegans, with minimal impact on the animal's fitness due to its robust innate immune response. This study aimed to understand the interactions between C. elegans and OrV by tracking the infection's progression during larval development. Four distinct stages of infection were identified on the basis of viral load, with a peak in capsid-encoding RNA2 coinciding with the first signs of viral egression. Transcriptomic analysis revealed temporal changes in gene expression and functions induced by the infection. A specific set of up-regulated genes remained active throughout the infection, and genes correlated and anticorrelated with virus accumulation were identified. Responses to OrV mirrored reactions to other biotic stressors, distinguishing between virus-specific responses and broader immune responses. Moreover, mutants of early response genes and defense-related processes showed altered viral load progression, uncovering additional players in the antiviral defense response.
Collapse
Affiliation(s)
- Victoria G. Castiglioni
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
| | - María J. Olmo-Uceda
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
| | - Ana Villena-Giménez
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
| | - Juan C. Muñoz-Sánchez
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
| | - Esmeralda G. Legarda
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
| | - Santiago F. Elena
- Institute of Integrative Systems Biology (I2SysBio), CSIC-Universitat de València, Paterna, 46980 Valencia, Spain
- Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
9
|
Nair T, Weathers BA, Stuhr NL, Nhan JD, Curran SP. Serotonin deficiency from constitutive SKN-1 activation drives pathogen apathy. Nat Commun 2024; 15:8129. [PMID: 39285192 PMCID: PMC11405893 DOI: 10.1038/s41467-024-52233-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/29/2024] [Indexed: 09/20/2024] Open
Abstract
When an organism encounters a pathogen, the host innate immune system activates to defend against pathogen colonization and toxic xenobiotics produced. C. elegans employ multiple defense systems to ensure survival when exposed to Pseudomonas aeruginosa including activation of the cytoprotective transcription factor SKN-1/NRF2. Although wildtype C. elegans quickly learn to avoid pathogens, here we describe a peculiar apathy-like behavior towards PA14 in animals with constitutive activation of SKN-1, whereby animals choose not to leave and continue to feed on the pathogen even when a non-pathogenic and healthspan-promoting food option is available. Although lacking the urgency to escape the infectious environment, animals with constitutive SKN-1 activity are not oblivious to the presence of the pathogen and display the typical pathogen-induced intestinal distension and eventual demise. SKN-1 activation, specifically in neurons and intestinal tissues, orchestrates a unique transcriptional program which leads to defects in serotonin signaling that is required from both neurons and non-neuronal tissues. Serotonin depletion from SKN-1 activation limits pathogen defenses capacity, drives the pathogen-associated apathy behaviors and induces a synthetic sensitivity to selective serotonin reuptake inhibitors. Taken together, our work reveals interesting insights into how animals perceive environmental pathogens and subsequently alter behavior and cellular programs to promote survival.
Collapse
Affiliation(s)
- Tripti Nair
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Brandy A Weathers
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Nicole L Stuhr
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - James D Nhan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Sean P Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Gonzalez X, Irazoqui JE. Distinct members of the Caenorhabditis elegans CeMbio reference microbiota exert cryptic virulence that is masked by host defense. Mol Microbiol 2024; 122:387-402. [PMID: 38623070 PMCID: PMC11480257 DOI: 10.1111/mmi.15258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/17/2024]
Abstract
Microbiotas are complex microbial communities that colonize specific niches in the host and provide essential organismal functions that are important in health and disease. Understanding the ability of each distinct community member to promote or impair host health, alone or in the context of the community, is imperative for understanding how differences in community structure affect host health and vice versa. Recently, a reference 12-member microbiota for the model organism Caenorhabditis elegans, known as CeMbio, was defined. Here, we show the differential ability of each CeMbio bacterial species to activate innate immunity through the conserved PMK-1/p38 MAPK, ACh-WNT, and HLH-30/TFEB pathways. Although distinct CeMbio members differed in their ability to activate the PMK-1/p38 pathway, the ability to do so did not correlate with bacterial-induced lifespan reduction in wild-type or immunodeficient animals. In contrast, most species activated HLH-30/TFEB and showed virulence toward hlh-30-deficient animals. These results suggest that the microbiota of C. elegans is rife with bacteria that can shorten the host's lifespan if host defense is compromised and that HLH-30/TFEB is a fundamental and key host protective factor.
Collapse
Affiliation(s)
- Xavier Gonzalez
- Immunology and Microbiology graduate program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester MA 01605
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester MA 01605
| | - Javier E. Irazoqui
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester MA 01605
| |
Collapse
|
11
|
Hajdú G, Szathmári C, Sőti C. Modeling Host-Pathogen Interactions in C. elegans: Lessons Learned from Pseudomonas aeruginosa Infection. Int J Mol Sci 2024; 25:7034. [PMID: 39000143 PMCID: PMC11241598 DOI: 10.3390/ijms25137034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Infections, such as that by the multiresistant opportunistic bacterial pathogen Pseudomonas aeruginosa, may pose a serious health risk, especially on vulnerable patient populations. The nematode Caenorhabditis elegans provides a simple organismal model to investigate both pathogenic mechanisms and the emerging role of innate immunity in host protection. Here, we review the virulence and infection strategies of P. aeruginosa and host defenses of C. elegans. We summarize the recognition mechanisms of patterns of pathogenesis, including novel pathogen-associated molecular patterns and surveillance immunity of translation, mitochondria, and lysosome-related organelles. We also review the regulation of antimicrobial and behavioral defenses by the worm's neuroendocrine system. We focus on how discoveries in this rich field align with well-characterized evolutionary conserved protective pathways, as well as on potential crossovers to human pathogenesis and innate immune responses.
Collapse
Affiliation(s)
- Gábor Hajdú
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csenge Szathmári
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
12
|
Kingsley SF, Seo Y, Wood A, Wani KA, Gonzalez X, Irazoqui J, Finkel SE, Tissenbaum HA. Glucose-fed microbiota alters C. elegans intestinal epithelium and increases susceptibility to multiple bacterial pathogens. Sci Rep 2024; 14:13177. [PMID: 38849503 PMCID: PMC11161463 DOI: 10.1038/s41598-024-63514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Overconsumption of dietary sugar can lead to many negative health effects including the development of Type 2 diabetes, metabolic syndrome, cardiovascular disease, and neurodegenerative disorders. Recently, the human intestinal microbiota, strongly associated with our overall health, has also been known to be affected by diet. However, mechanistic insight into the importance of the human intestinal microbiota and the effects of chronic sugar ingestion has not been possible largely due to the complexity of the human microbiome which contains hundreds of types of organisms. Here, we use an interspecies C. elegans/E. coli system, where E. coli are subjected to high sugar, then consumed by the bacterivore host C. elegans to become the microbiota. This glucose-fed microbiota results in a significant lifespan reduction accompanied by reduced healthspan (locomotion), reduced stress resistance, and changes in behavior and feeding. Lifespan reduction is also accompanied by two potential major contributors: increased intestinal bacterial density and increased concentration of reactive oxygen species. The glucose-fed microbiota accelerated the age-related development of intestinal cell permeability, intestinal distention, and dysregulation of immune effectors. Ultimately, the changes in the intestinal epithelium due to aging with the glucose-fed microbiota results in increased susceptibility to multiple bacterial pathogens. Taken together, our data reveal that chronic ingestion of sugar, such as a Western diet, has profound health effects on the host due to changes in the microbiota and may contribute to the current increased incidence of ailments including inflammatory bowel diseases as well as multiple age-related diseases.
Collapse
Affiliation(s)
- Samuel F Kingsley
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Yonghak Seo
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Alicia Wood
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Khursheed A Wani
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Xavier Gonzalez
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Javier Irazoqui
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Steven E Finkel
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089-2910, USA
| | - Heidi A Tissenbaum
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
13
|
Pu X, Qi B. Lysosomal dysfunction by inactivation of V-ATPase drives innate immune response in C. elegans. Cell Rep 2024; 43:114138. [PMID: 38678555 DOI: 10.1016/j.celrep.2024.114138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024] Open
Abstract
Pathogens target vacuolar ATPase (V-ATPase) to inhibit lysosomal acidification or lysosomal fusion, causing lysosomal dysfunction. However, it remains unknown whether cells can detect dysfunctional lysosomes and initiate an immune response. In this study, we discover that dysfunction of lysosomes caused by inactivation of V-ATPase enhances innate immunity against bacterial infections. We find that lysosomal V-ATPase interacts with DVE-1, whose nuclear localization serves as a proxy for the induction of mitochondrial unfolded protein response (UPRmt). The inactivation of V-ATPase promotes the nuclear localization of DVE-1, activating UPRmt and inducing downstream immune response genes. Furthermore, pathogen resistance conferred by inactivation of V-ATPase requires dve-1 and its downstream immune effectors. Interestingly, animals grow slower after vha RNAi, suggesting that the vha-RNAi-induced immune response costs the most energy through activation of DVE-1, which trades off with growth. This study reveals how dysfunctional lysosomes can trigger an immune response, emphasizing the importance of conserving energy during immune defense.
Collapse
Affiliation(s)
- Xuepiao Pu
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Bin Qi
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| |
Collapse
|
14
|
Torzone SK, Breen PC, Cohen NR, Simmons KN, Dowen RH. The TWK-26 potassium channel governs nutrient absorption in the C. elegans intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592787. [PMID: 38766028 PMCID: PMC11100751 DOI: 10.1101/2024.05.06.592787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Ion channels are necessary for proper water and nutrient absorption in the intestine, which supports cellular metabolism and organismal growth. While a role for Na + co-transporters and pumps in intestinal nutrient absorption is well defined, how individual K + uniporters function to maintain ion homeostasis is poorly understood. Using Caenorhabditis elegans , we show that a gain-of-function mutation in twk-26 , which encodes a two-pore domain K + ion channel orthologous to human KCNK3, facilitates nutrient absorption and suppresses the metabolic and developmental defects displayed by impaired intestinal MAP Kinase (MAPK) signaling. Mutations in drl-1 and flr-4, which encode two components of this MAPK pathway, cause severe growth defects, reduced lipid storage, and a dramatic increase in autophagic lysosomes, which mirror dietary restriction phenotypes. Additionally, these MAPK mutants display structural defects of the intestine and an impaired defecation motor program. We find that activation of TWK-26 reverses the dietary restriction-like state of the MAPK mutants by restoring intestinal nutrient absorption without correcting the intestinal bloating or defecation defects. This study provides unique insight into the mechanisms by which intestinal K + ion channels support intestinal metabolic homeostasis.
Collapse
|
15
|
Kywe C, Lundquist EA, Ackley BD, Lansdon P. The MAB-5/Hox family transcription factor is important for Caenorhabditis elegans innate immune response to Staphylococcus epidermidis infection. G3 (BETHESDA, MD.) 2024; 14:jkae054. [PMID: 38478633 PMCID: PMC11075571 DOI: 10.1093/g3journal/jkae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/17/2024] [Accepted: 03/03/2024] [Indexed: 04/12/2024]
Abstract
Innate immunity functions as a rapid defense against broad classes of pathogenic agents. While the mechanisms of innate immunity in response to antigen exposure are well-studied, how pathogen exposure activates the innate immune responses and the role of genetic variation in immune activity is currently being investigated. Previously, we showed significant survival differences between the N2 and the CB4856 Caenorhabditis elegans isolates in response to Staphylococcus epidermidis infection. One of those differences was expression of the mab-5 Hox family transcription factor, which was induced in N2, but not CB4856, after infection. In this study, we use survival assays and RNA-sequencing to better understand the role of mab-5 in response to S. epidermidis. We found that mab-5 loss-of-function (LOF) mutants were more susceptible to S. epidermidis infection than N2 or mab-5 gain-of-function (GOF) mutants, but not as susceptible as CB4856 animals. We then conducted transcriptome analysis of infected worms and found considerable differences in gene expression profiles when comparing animals with mab-5 LOF to either N2 or mab-5 GOF. N2 and mab-5 GOF animals showed a significant enrichment in expression of immune genes and C-type lectins, whereas mab-5 LOF mutants did not. Overall, gene expression profiling in mab-5 mutants provided insight into MAB-5 regulation of the transcriptomic response of C. elegans to pathogenic bacteria and helps us to understand mechanisms of innate immune activation and the role that transcriptional regulation plays in organismal health.
Collapse
Affiliation(s)
- Christopher Kywe
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Erik A Lundquist
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Brian D Ackley
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Patrick Lansdon
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
16
|
González R, Félix MA. Caenorhabditis elegans immune responses to microsporidia and viruses. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 154:105148. [PMID: 38325500 DOI: 10.1016/j.dci.2024.105148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
The model organism Caenorhabditis elegans is susceptible to infection by obligate intracellular pathogens, specifically microsporidia and viruses. These intracellular pathogens infect intestinal cells, or, for some microsporidia, epidermal cells. Strikingly, intestinal cell infections by viruses or microsporidia trigger a common transcriptional response, activated in part by the ZIP-1 transcription factor. Among the strongest activated genes in this response are ubiquitin-pathway members and members of the pals family, an intriguing gene family with cross-regulations of different members of genomic clusters. Some of the induced genes participate in host defense against the pathogens, for example through ubiquitin-mediated inhibition. Other mechanisms defend the host specifically against viral infections, including antiviral RNA interference and uridylation. These various immune responses are altered by environmental factors and by intraspecific genetic variation of the host. These pathogens were first isolated 15 years ago and much remains to be discovered using C. elegans genetics; also, other intracellular pathogens of C. elegans may yet to be discovered.
Collapse
Affiliation(s)
- Rubén González
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France.
| | - Marie-Anne Félix
- Institut de Biologie de l'École Normale Supérieure, CNRS, INSERM, 75005, Paris, France
| |
Collapse
|
17
|
El Jarkass HT, Reinke AW. Pathogen evolution: Protective microbes act as a double-edged sword. Curr Biol 2024; 34:R247-R249. [PMID: 38531318 DOI: 10.1016/j.cub.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Vaccines and infection can sometimes cause incomplete immunity, which allows for pathogen re-infection with decreased disease severity but also contributes to the evolution of pathogen virulence. A new study demonstrates that incomplete immunity from resident protective microbes results in similar evolutionary trajectories.
Collapse
Affiliation(s)
| | - Aaron W Reinke
- Department of Molecular Genetics, University of Toronto, Toronto ON M5G 1M1, Canada.
| |
Collapse
|
18
|
Hoang KL, Read TD, King KC. Incomplete immunity in a natural animal-microbiota interaction selects for higher pathogen virulence. Curr Biol 2024; 34:1357-1363.e3. [PMID: 38430909 PMCID: PMC10962313 DOI: 10.1016/j.cub.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024]
Abstract
Incomplete immunity in recovered hosts is predicted to favor more virulent pathogens upon re-infection in the population.1 The microbiota colonizing animals can generate a similarly long-lasting, partial immune response, allowing for infection but dampened disease severity.2 We tracked the evolutionary trajectories of a widespread pathogen (Pseudomonas aeruginosa), experimentally passaged through populations of nematodes immune-primed by a natural microbiota member (P. berkeleyensis). This bacterium can induce genes regulated by a mitogen-activated protein kinase (MAPK) signaling pathway effective at conferring protection against pathogen-induced death despite infection.3 Across host populations, this incomplete immunity selected for pathogens more than twice as likely to kill as those evolved in non-primed (i.e., naive) or immune-compromised (mutants with a knockout of the MAPK ortholog) control populations. Despite the higher virulence, pathogen molecular evolution in immune-primed hosts was slow and constrained. In comparison, evolving pathogens in immune-compromised hosts were characterized by substantial genomic differentiation and attenuated virulence. These findings directly attribute the incomplete host immunity induced from microbiota as a significant force shaping the virulence and evolutionary dynamics of novel infectious diseases.
Collapse
Affiliation(s)
- Kim L Hoang
- Department of Biology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK; Division of Infectious Diseases, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA.
| | - Timothy D Read
- Division of Infectious Diseases, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Kayla C King
- Department of Biology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK; Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, BC V6T 1Z4, Canada; Department of Microbiology & Immunology, University of British Columbia, 1365 - 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
19
|
Matsuda A, Ishida T, Tanimoto Y, Wada T, Kage-Nakadai E. Pathogenicity of enterotoxigenic Escherichia coli in Caenorhabditis elegans as an alternative model host. Biosci Biotechnol Biochem 2024; 88:453-459. [PMID: 38159930 DOI: 10.1093/bbb/zbad185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC), one of the diarrheagenic E. coli, is the most common cause of diarrhea in developing country and in travelers to those areas. In this study, Caenorhabditis elegans was used as an alternative model host to evaluate ETEC infections. The ETEC strain ETEC1, which was isolated from a patient with diarrhea, possessed enterotoxins STh, LT1, and EAST1 and colonization factors CS2 and CS3. Live ETEC1 shortened the life span and body size of C. elegans in association with increased expression of enterotoxin genes and intestinal colonization. In contrast, heat-killed ETEC1 did not affect the life span of C. elegans. Caenorhabditis elegans infected with ETEC1 showed upregulated expression of genes related to insulin-like peptides and host defense responses. These results suggest that ETEC1 exhibits pathogenicity through intestinal colonization and enterotoxin production in C. elegans. This system is useful as an ETEC infection model.
Collapse
Affiliation(s)
- Anri Matsuda
- Graduate School of Human Life and Ecology, Osaka Metropolitan University, Osaka, Japan
| | - Takashi Ishida
- Corporate Strategy Department, Kyowa Hakko Bio Co., Ltd, Tokyo, Japan
| | - Yoshihiko Tanimoto
- Graduate School of Human Life and Ecology, Osaka Metropolitan University, Osaka, Japan
- Department of Infectious Diseases, Kobe Institute of Health, Hyogo, Japan
| | - Takayuki Wada
- Graduate School of Human Life and Ecology, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Eriko Kage-Nakadai
- Graduate School of Human Life and Ecology, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
20
|
Turner CD, Ramos CM, Curran SP. Disrupting the SKN-1 homeostat: mechanistic insights and phenotypic outcomes. FRONTIERS IN AGING 2024; 5:1369740. [PMID: 38501033 PMCID: PMC10944932 DOI: 10.3389/fragi.2024.1369740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/15/2024] [Indexed: 03/20/2024]
Abstract
The mechanisms that govern maintenance of cellular homeostasis are crucial to the lifespan and healthspan of all living systems. As an organism ages, there is a gradual decline in cellular homeostasis that leads to senescence and death. As an organism lives into advanced age, the cells within will attempt to abate age-related decline by enhancing the activity of cellular stress pathways. The regulation of cellular stress responses by transcription factors SKN-1/Nrf2 is a well characterized pathway in which cellular stress, particularly xenobiotic stress, is abated by SKN-1/Nrf2-mediated transcriptional activation of the Phase II detoxification pathway. However, SKN-1/Nrf2 also regulates a multitude of other processes including development, pathogenic stress responses, proteostasis, and lipid metabolism. While this process is typically tightly regulated, constitutive activation of SKN-1/Nrf2 is detrimental to organismal health, this raises interesting questions surrounding the tradeoff between SKN-1/Nrf2 cryoprotection and cellular health and the ability of cells to deactivate stress response pathways post stress. Recent work has determined that transcriptional programs of SKN-1 can be redirected or suppressed to abate negative health outcomes of constitutive activation. Here we will detail the mechanisms by which SKN-1 is controlled, which are important for our understanding of SKN-1/Nrf2 cytoprotection across the lifespan.
Collapse
Affiliation(s)
- Chris D. Turner
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Carmen M. Ramos
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
- Dornsife College of Letters, Arts, and Sciences, Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, United States
| | - Sean P. Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
21
|
Sengupta T, St. Ange J, Kaletsky R, Moore RS, Seto RJ, Marogi J, Myhrvold C, Gitai Z, Murphy CT. A natural bacterial pathogen of C. elegans uses a small RNA to induce transgenerational inheritance of learned avoidance. PLoS Genet 2024; 20:e1011178. [PMID: 38547071 PMCID: PMC10977744 DOI: 10.1371/journal.pgen.1011178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/09/2024] [Indexed: 04/02/2024] Open
Abstract
C. elegans can learn to avoid pathogenic bacteria through several mechanisms, including bacterial small RNA-induced learned avoidance behavior, which can be inherited transgenerationally. Previously, we discovered that a small RNA from a clinical isolate of Pseudomonas aeruginosa, PA14, induces learned avoidance and transgenerational inheritance of that avoidance in C. elegans. Pseudomonas aeruginosa is an important human pathogen, and there are other Pseudomonads in C. elegans' natural habitat, but it is unclear whether C. elegans ever encounters PA14-like bacteria in the wild. Thus, it is not known if small RNAs from bacteria found in C. elegans' natural habitat can also regulate host behavior and produce heritable behavioral effects. Here we screened a set of wild habitat bacteria, and found that a pathogenic Pseudomonas vranovensis strain isolated from the C. elegans microbiota, GRb0427, regulates worm behavior: worms learn to avoid this pathogenic bacterium following exposure, and this learned avoidance is inherited for four generations. The learned response is entirely mediated by bacterially-produced small RNAs, which induce avoidance and transgenerational inheritance, providing further support that such mechanisms of learning and inheritance exist in the wild. We identified Pv1, a small RNA expressed in P. vranovensis, that has a 16-nucleotide match to an exon of the C. elegans gene maco-1. Pv1 is both necessary and sufficient to induce learned avoidance of Grb0427. However, Pv1 also results in avoidance of a beneficial microbiome strain, P. mendocina. Our findings suggest that bacterial small RNA-mediated regulation of host behavior and its transgenerational inheritance may be functional in C. elegans' natural environment, and that this potentially maladaptive response may favor reversal of the transgenerational memory after a few generations. Our data also suggest that different bacterial small RNA-mediated regulation systems evolved independently, but define shared molecular features of bacterial small RNAs that produce transgenerationally-inherited effects.
Collapse
Affiliation(s)
- Titas Sengupta
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Jonathan St. Ange
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Rachel Kaletsky
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Rebecca S. Moore
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Renee J. Seto
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Jacob Marogi
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Zemer Gitai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Coleen T. Murphy
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|
22
|
Dhakal A, Salim C, Skelly M, Amichan Y, Lamm AT, Hundley HA. ADARs regulate cuticle collagen expression and promote survival to pathogen infection. BMC Biol 2024; 22:37. [PMID: 38360623 PMCID: PMC10870475 DOI: 10.1186/s12915-024-01840-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND In all organisms, the innate immune system defends against pathogens through basal expression of molecules that provide critical barriers to invasion and inducible expression of effectors that combat infection. The adenosine deaminase that act on RNA (ADAR) family of RNA-binding proteins has been reported to influence innate immunity in metazoans. However, studies on the susceptibility of ADAR mutant animals to infection are largely lacking. RESULTS Here, by analyzing adr-1 and adr-2 null mutants in well-established slow-killing assays, we find that both Caenorhabditis elegans ADARs are important for organismal survival to gram-negative and gram-positive bacteria, all of which are pathogenic to humans. Furthermore, our high-throughput sequencing and genetic analysis reveal that ADR-1 and ADR-2 function in the same pathway to regulate collagen expression. Consistent with this finding, our scanning electron microscopy studies indicate adr-1;adr-2 mutant animals also have altered cuticle morphology prior to pathogen exposure. CONCLUSIONS Our data uncover a critical role of the C. elegans ADAR family of RNA-binding proteins in promoting cuticular collagen expression, which represents a new post-transcriptional regulatory node that influences the extracellular matrix. In addition, we provide the first evidence that ADAR mutant animals have altered susceptibility to infection with several opportunistic human pathogens, suggesting a broader role of ADARs in altering physical barriers to infection to influence innate immunity.
Collapse
Affiliation(s)
- Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine-Bloomington, Bloomington, IN, 47405, USA
| | - Chinnu Salim
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Mary Skelly
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Yarden Amichan
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Ayelet T Lamm
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Heather A Hundley
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
23
|
Tran TD, Luallen RJ. An organismal understanding of C. elegans innate immune responses, from pathogen recognition to multigenerational resistance. Semin Cell Dev Biol 2024; 154:77-84. [PMID: 36966075 PMCID: PMC10517082 DOI: 10.1016/j.semcdb.2023.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 03/27/2023]
Abstract
The nematode Caenorhabditis elegans has been a model for studying infection since the early 2000s and many major discoveries have been made regarding its innate immune responses. C. elegans has been found to utilize some key conserved aspects of immune responses and signaling, but new interesting features of innate immunity have also been discovered in the organism that might have broader implications in higher eukaryotes such as mammals. Some of the distinctive features of C. elegans innate immunity involve the mechanisms this bacterivore uses to detect infection and mount specific immune responses to different pathogens, despite lacking putative orthologs of many important innate immune components, including cellular immunity, the inflammasome, complement, or melanization. Even when orthologs of known immune factors exist, there appears to be an absence of canonical functions, most notably the lack of pattern recognition by its sole Toll-like receptor. Instead, recent research suggests that C. elegans senses infection by specific pathogens through contextual information, including unique products produced by the pathogen or infection-induced disruption of host physiology, similar to the proposed detection of patterns of pathogenesis in mammalian systems. Interestingly, C. elegans can also transfer information of past infection to their progeny, providing robust protection for their offspring in face of persisting pathogens, in part through the RNAi pathway as well as potential new mechanisms that remain to be elucidated. Altogether, some of these strategies employed by C. elegans share key conceptual features with vertebrate adaptive immunity, as the animal can differentiate specific microbial features, as well as propagate a form of immune memory to their offspring.
Collapse
Affiliation(s)
- Tuan D Tran
- Department of Biology San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - Robert J Luallen
- Department of Biology San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| |
Collapse
|
24
|
Nair T, Weathers BA, Stuhr NL, Nhan JD, Curran SP. Serotonin deficiency from constitutive SKN-1 activation drives pathogen apathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.579755. [PMID: 38405962 PMCID: PMC10888766 DOI: 10.1101/2024.02.10.579755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
When an organism encounters a pathogen, the host innate immune system activates to defend against pathogen colonization and toxic xenobiotics produced. C. elegans employ multiple defense systems to ensure survival when exposed to Pseudomonas aeruginosa including activation of the cytoprotective transcription factor SKN-1/NRF2. Although wildtype C. elegans quickly learn to avoid pathogens, here we describe a peculiar apathy-like behavior towards PA14 in animals with constitutive activation of SKN-1, whereby animals choose not to leave and continue to feed on the pathogen even when a non-pathogenic and healthspan-promoting food option is available. Although lacking the urgency to escape the infectious environment, animals with constitutive SKN-1 activity are not oblivious to the presence of the pathogen and display the typical pathogen-induced intestinal distension and eventual demise. SKN-1 activation, specifically in neurons and intestinal tissues, orchestrates a unique transcriptional program which leads to defects in serotonin signaling that is required from both neurons and non-neuronal tissues. Serotonin depletion from SKN-1 activation limits pathogen defense capacity, drives the pathogen-associated apathy behaviors and induces a synthetic sensitivity to selective serotonin reuptake inhibitors. Taken together, our work reveals new insights into how animals perceive environmental pathogens and subsequently alter behavior and cellular programs to promote survival. KEY POINTS Identify an apathy-like behavioral response for pathogens resulting from the constitutive activation of the cytoprotective transcription factor SKN-1.Uncover the obligate role for serotonin synthesis in both neuronal and non-neuronal cells for the apathy-like state and ability of serotonin treatment to restore normal behaviors.Characterize the timing and tissue specificity of SKN-1 nuclear localization in neurons and intestinal cells in response to pathogen exposure.Define the unique and context-specific transcriptional signatures of animals with constitutive SKN-1 activation when exposed to pathogenic environments.Reveal necessity for both neuronal and non-neuronal serotonin signaling in host survival from pathogen infection.
Collapse
|
25
|
Bollen DP, Reddy KC, Lascarez-Lagunas LI, Kim DH, Colaiácovo MP. Germline mitotic quiescence and cell death are induced in Caenorhabditis elegans by exposure to pathogenic Pseudomonas aeruginosa. Genetics 2024; 226:iyad197. [PMID: 37956057 PMCID: PMC10763535 DOI: 10.1093/genetics/iyad197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/08/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
The impact of exposure to microbial pathogens on animal reproductive capacity and germline physiology is not well understood. The nematode Caenorhabditis elegans is a bacterivore that encounters pathogenic microbes in its natural environment. How pathogenic bacteria affect host reproductive capacity of C. elegans is not well understood. Here, we show that exposure of C. elegans hermaphrodites to the Gram-negative pathogen Pseudomonas aeruginosa causes a marked reduction in brood size with concomitant reduction in the number of nuclei in the germline and gonad size. We define 2 processes that are induced that contribute to the decrease in the number of germ cell nuclei. First, we observe that infection with P. aeruginosa leads to the induction of germ cell apoptosis. Second, we observe that this exposure induces mitotic quiescence in the proliferative zone of the C. elegans gonad. Importantly, these processes appear to be reversible; when animals are removed from the presence of P. aeruginosa, germ cell apoptosis is abated, germ cell nuclei numbers increase, and brood sizes recover. The reversible germline dynamics during exposure to P. aeruginosa may represent an adaptive response to improve survival of progeny and may serve to facilitate resource allocation that promotes survival during pathogen infection.
Collapse
Affiliation(s)
- Daniel P Bollen
- Division of Infectious Diseases and Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kirthi C Reddy
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Dennis H Kim
- Division of Infectious Diseases and Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Monica P Colaiácovo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
González R, Félix MA. Naturally-associated bacteria modulate Orsay virus infection of Caenorhabditis elegans. PLoS Pathog 2024; 20:e1011947. [PMID: 38232128 PMCID: PMC10824439 DOI: 10.1371/journal.ppat.1011947] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 01/29/2024] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Microbes associated with an organism can significantly modulate its susceptibility to viral infections, but our understanding of the influence of individual microbes remains limited. The nematode Caenorhabditis elegans is a model organism that in nature inhabits environments rich in bacteria. Here, we examine the impact of 71 naturally associated bacteria on C. elegans susceptibility to its only known natural virus, the Orsay virus. Our findings reveal that viral infection of C. elegans is significantly influenced by monobacterial environments. Compared to an Escherichia coli environmental reference, the majority of tested bacteria reduced C. elegans susceptibility to viral infection. This reduction is not caused by virion degradation or poor animal nutrition by the bacteria. The repression of viral infection by the bacterial strains Chryseobacterium JUb44 and Sphingobacterium BIGb0172 does not require the RIG-I homolog DRH-1, which is known to activate antiviral responses such as RNA interference and transcriptional regulation. Our research highlights the necessity of considering natural biotic environments in viral infection studies and opens the way future research on host-microbe-virus interactions.
Collapse
Affiliation(s)
- Rubén González
- Institut de Biologie de l’École Normale Supérieure, CNRS, INSERM, Paris, France
| | - Marie-Anne Félix
- Institut de Biologie de l’École Normale Supérieure, CNRS, INSERM, Paris, France
| |
Collapse
|
27
|
Pradhan J, Pradhan D, Sahu JK, Mishra S, Mallick S, Das S, Negi VD. A novel rspA gene regulates biofilm formation and virulence of Salmonella Typhimurium. Microb Pathog 2023; 185:106432. [PMID: 37926364 DOI: 10.1016/j.micpath.2023.106432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Salmonella spp. are facultative anaerobic, Gram-negative, rod-shaped bacteria and belongs to the Enterobacteriaceae family. Although much has been known about Salmonella pathogenesis, the functional characterizations of certain genes are yet to be explored. The rspA (STM14_1818) is one such gene with putative dehydratase function, and its role in pathogenesis is unknown. The background information showed that rspA gene is upregulated in Salmonella when it resides inside macrophages, which led us to investigate its role in Salmonella pathogenesis. We generated the rspA knockout strain and complement strain in S. Typhimurium 14028. Ex-vivo and in-vivo infectivity was looked at macrophage and epithelial cell lines and Caenorhabditis elegans (C. elegans). The mutant strain differentially formed the biofilm at different temperatures by altering the expression of genes involved in the synthesis of cellulose and curli. Besides, the mutant strain is hyperproliferative intracellularly and showed increased bacterial burden in C. elegans. The mutant strain became more infectious and lethal, causing faster death of the worms than the wild type, and also modulates the worm's innate immunity. Thus, we found that the rspA deletion mutant was more pathogenic. In this study, we concluded that the rspA gene differentially regulates the biofilm formation in a temperature dependent manner by modulating the genes involved in the synthesis of cellulose and curli and negatively regulates the Salmonella virulence for longer persistence inside the host.
Collapse
Affiliation(s)
- Jasmin Pradhan
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Diana Pradhan
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Jugal Kishor Sahu
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Satyajit Mishra
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Swarupa Mallick
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Surajit Das
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Vidya Devi Negi
- Laboratory of Infection Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
28
|
Yang RQ, Chen YH, Wu QY, Tang J, Niu SZ, Zhao Q, Ma YC, Zou CG. Indole produced during dysbiosis mediates host-microorganism chemical communication. eLife 2023; 12:e85362. [PMID: 37987602 PMCID: PMC10691800 DOI: 10.7554/elife.85362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
An imbalance of the gut microbiota, termed dysbiosis, has a substantial impact on host physiology. However, the mechanism by which host deals with gut dysbiosis to maintain fitness remains largely unknown. In Caenorhabditis elegans, Escherichia coli, which is its bacterial diet, proliferates in its intestinal lumen during aging. Here, we demonstrate that progressive intestinal proliferation of E. coli activates the transcription factor DAF-16, which is required for maintenance of longevity and organismal fitness in worms with age. DAF-16 up-regulates two lysozymes lys-7 and lys-8, thus limiting the bacterial accumulation in the gut of worms during aging. During dysbiosis, the levels of indole produced by E. coli are increased in worms. Indole is involved in the activation of DAF-16 by TRPA-1 in neurons of worms. Our finding demonstrates that indole functions as a microbial signal of gut dysbiosis to promote fitness of the host.
Collapse
Affiliation(s)
- Rui-Qiu Yang
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| | - Yong-Hong Chen
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| | - Qin-yi Wu
- Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Yunnan University of Traditional Chinese MedicineKunmingChina
| | - Jie Tang
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
- Yunnan Key Laboratory of Vaccine Research Development on Severe Infectious Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Shan-Zhuang Niu
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| | - Qiu Zhao
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| | - Yi-Cheng Ma
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| | - Cheng-Gang Zou
- State key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan UniversityKunmingChina
| |
Collapse
|
29
|
Gonzalez X, Irazoqui JE. Distinct members of the C. elegans CeMbio reference microbiota exert cryptic virulence and infection protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565327. [PMID: 37961109 PMCID: PMC10635080 DOI: 10.1101/2023.11.02.565327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Microbiotas are complex microbial communities that colonize specific niches in the host and provide essential organismal functions that are important in health and disease. A key aspect is the ability of each distinct community member to promote or impair host health, alone or in the context of the community, in hosts with varied levels of immune competence. Understanding such interactions is limited by the complexity and experimental accessibility of current systems and models. Recently, a reference twelve-member microbiota for the model organism C. elegans, known as CeMbio, was defined to aid the dissection of conserved host-microbiota interactions. Understanding the physiological impact of the CeMbio bacteria on C. elegans is in its infancy. Here, we show the differential ability of each CeMbio bacterial species to activate innate immunity through the conserved PMK-1/p38 MAPK, ACh/WNT, and HLH-30/TFEB pathways. Using immunodeficient animals, we uncovered several examples of bacterial 'cryptic' virulence, or virulence that was masked by the host defense response. The ability to activate the PMK-1/p38 pathway did not correlate with bacterial virulence in wild type or immunodeficient animals. In contrast, ten out of twelve species activated HLH-30/TFEB, and most showed virulence towards hlh-30-deficient animals. In addition, we identified Pseudomonas lurida as a pathogen in wild type animals, and Acinetobacter guillouiae as avirulent despite activating all three pathways. Moreover, short pre-exposure to A. guillouiae promoted host survival of infection with P. lurida, which was dependent on PMK-1/p38 MAPK and HLH-30/TFEB. These results suggest that the microbiota of C. elegans is rife with "opportunistic" pathogens, and that HLH-30/TFEB is a fundamental and key host protective factor. Furthermore, they support the idea that bacteria like A. guillouiae evolved the ability to induce host innate immunity to improve host fitness when confronted with pathogens, providing new insights into how colonization order impacts host health.
Collapse
Affiliation(s)
- Xavier Gonzalez
- Immunology and Microbiology graduate program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester MA 01605
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester MA 01605
| | - Javier E. Irazoqui
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester MA 01605
| |
Collapse
|
30
|
Sengupta T, St. Ange J, Moore R, Kaletsky R, Marogi J, Myhrvold C, Gitai Z, Murphy CT. A natural bacterial pathogen of C. elegans uses a small RNA to induce transgenerational inheritance of learned avoidance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549962. [PMID: 37503135 PMCID: PMC10370180 DOI: 10.1101/2023.07.20.549962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Previously, we discovered that a small RNA from a clinical isolate of Pseudomonas aeruginosa, PA14, induces learned avoidance and its transgenerational inheritance in C. elegans. Pseudomonas aeruginosa is an important human pathogen, and there are other Pseudomonads in C. elegans' natural habitat, but it is unclear whether C. elegans ever encounters PA14-like bacteria in the wild. Thus, it is not known if small RNAs from bacteria found in C. elegans' natural habitat can also regulate host behavior and produce heritable behavioral effects. Here we found that a pathogenic Pseudomonas vranovensis strain isolated from the C. elegans microbiota, GRb0427, like PA14, regulates worm behavior: worms learn to avoid this pathogenic bacterium following exposure to GRb0427, and this learned avoidance is inherited for four generations. The learned response is entirely mediated by bacterially-produced small RNAs, which induce avoidance and transgenerational inheritance, providing further support that such mechanisms of learning and inheritance exist in the wild. Using bacterial small RNA sequencing, we identified Pv1, a small RNA from GRb0427, that matches the sequence of C. elegans maco-1. We find that Pv1 is both necessary and sufficient to induce learned avoidance of Grb0427. However, Pv1 also results in avoidance of a beneficial microbiome strain, P. mendocina; this potentially maladaptive response may favor reversal of the transgenerational memory after a few generations. Our findings suggest that bacterial small RNA-mediated regulation of host behavior and its transgenerational inheritance are functional in C. elegans' natural environment, and that different bacterial small RNA-mediated regulation systems evolved independently but define shared molecular features of bacterial small RNAs that produce transgenerationally-inherited effects.
Collapse
Affiliation(s)
- Titas Sengupta
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jonathan St. Ange
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rebecca Moore
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rachel Kaletsky
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jacob Marogi
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Zemer Gitai
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T. Murphy
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
31
|
Roy S, Kc HR, Roberts J, Hastings J, Gilmore DF, Shields RC, Alam MA. Development and Antibacterial Properties of 4-[4-(Anilinomethyl)-3-phenylpyrazol-1-yl]benzoic Acid Derivatives as Fatty Acid Biosynthesis Inhibitors. J Med Chem 2023; 66:13622-13645. [PMID: 37729113 PMCID: PMC10591900 DOI: 10.1021/acs.jmedchem.3c00969] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
A number of novel pyrazole derivatives have been synthesized, and several of these compounds are potent antibacterial agents with minimum inhibitory concentrations as low as 0.5 μg/mL. Human cell lines were tolerant to these lead compounds, and they showed negligible hemolytic effects at high concentrations. These bactericidal compounds are very effective against bacterial growth in both planktonic and biofilm contexts. Various techniques were applied to show the inhibition of biofilm growth and eradication of preformed biofilms by lead compounds. Potent compounds are more effective against persisters than positive controls. In vivo studies revealed that lead compounds are effective in rescuing C. elegans from bacterial infections. Several methods were applied to determine the mode of action including membrane permeability assay and SEM micrograph studies. Furthermore, CRISPRi studies led to the determination of these compounds as fatty acid biosynthesis (FAB) inhibitors.
Collapse
Affiliation(s)
- Subrata Roy
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - Hansa Raj Kc
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - Justin Roberts
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - Jared Hastings
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - David F Gilmore
- Department of Biological Sciences, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - Robert C Shields
- Department of Biological Sciences, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| | - Mohammad A Alam
- Department of Chemistry and Physics, College of Science and Mathematics, Arkansas State University, Jonesboro, Arkansas 72467, United States
| |
Collapse
|
32
|
Zhang H, Zeng W, Zhao MM, Wang J, Wang Q, Chen T, Zhang Y, Lee W, Chen S, Zhang Y, Lan X, Xiang Y. Caenorhabditis elegans LIN-24, a homolog of bacterial pore-forming toxin, protects the host from microbial infection. FASEB J 2023; 37:e23162. [PMID: 37682220 DOI: 10.1096/fj.202300063r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/18/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023]
Abstract
Aerolysin-like pore-forming protein (af-PFP) superfamily members are double-edge swords that assist the bacterial infection but shied bacteria from the host by various mechanisms in some species including the toad Bombina maxima and zebrafish. While members of this family are widely expressed in all kingdoms, especially non-bacteria species, it remains unclear whether their anti-bacterial function is conserved. LIN-24 is an af-PFP that is constitutively expressed throughout the Caenorhabditis elegans lifespan. Here, we observed that LIN-24 knockdown reduced the maximum lifespan of worms. RNA-seq analysis identified 323 differentially expressed genes (DEGs) post-LIN-24 knockdown that were enriched in "immune response" and "lysosome pathway," suggesting a possible role for LIN-24 in resisting microbial infection. In line with this, we found that Pseudomonas aeruginosa 14 (PA14) infection induced LIN-24 expression, and that survival after PA14 infection was significantly reduced by LIN-24 knockdown. In contrast, LIN-24 overexpression (LIN-24-OE) conferred protection against PA14 infection, with worms showing longer survival time and reduced bacterial load. Weighted gene co-expression network analysis of LIN-24-OE worms showed that the highest correlation module was enriched in factors related to immunity and the defense response. Finally, by predicting transcription factors from RNA-seq data and knocking down candidate transcription factors in LIN-24-OE worms, we revealed that LIN-24 may protect worms against bacterial infection by stimulating DAF-16-mediated immune responses. These findings agree with our previous studies showing an anti-microbial role for the amphibian-derived af-PFP complex βγ-CAT, suggesting that af-PFPs may play a conserved role in combatting microbial infections. Further research is needed to determine the roles this protein family plays in other physio-pathological processes, such as metabolism, longevity, and aging.
Collapse
Affiliation(s)
- Huijie Zhang
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Weirong Zeng
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Ming-Ming Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Jiali Wang
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Qiquan Wang
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Ting Chen
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Yuyan Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Wenhui Lee
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Shenghan Chen
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Xinqiang Lan
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Yang Xiang
- Metabolic Control and Aging, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| |
Collapse
|
33
|
Mokoena NZ, Steyn H, Hugo A, Dix-Peek T, Dickens C, Gcilitshana OMN, Sebolai O, Albertyn J, Pohl CH. Eicosapentaenoic acid influences the pathogenesis of Candida albicans in Caenorhabditis elegans via inhibition of hyphal formation and stimulation of the host immune response. Med Microbiol Immunol 2023; 212:349-368. [PMID: 37672050 PMCID: PMC10501937 DOI: 10.1007/s00430-023-00777-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 08/18/2023] [Indexed: 09/07/2023]
Abstract
The intake of omega-3 polyunsaturated fatty acids, including eicosapentaenoic acid (EPA), is associated with health benefits due to its anti-inflammatory properties. This fatty acid also exhibits antifungal properties in vitro. In order to determine if this antifungal property is valid in vivo, we examined how EPA affects Candida albicans pathogenesis in the Caenorhabditis elegans infection model, an alternative to mammalian host models. The nematodes were supplemented with EPA prior to infection, and the influence of EPA on C. elegans lipid metabolism, survival and immune response was studied. In addition, the influence of EPA on hyphal formation in C. albicans was investigated. It was discovered that EPA supplementation changed the lipid composition, but not the unsaturation index of C. elegans by regulating genes involved in fatty acid and eicosanoid production. EPA supplementation also delayed killing of C. elegans by C. albicans due to the inhibition of hyphal formation in vivo, via the action of the eicosanoid metabolite of EPA, 17,18-epoxyeicosatetraenoic acid. Moreover, EPA supplementation also caused differential expression of biofilm-related gene expression in C. albicans and stimulated the immune response of C. elegans. This provides a link between EPA and host susceptibility to microbial infection in this model.
Collapse
Affiliation(s)
- N Z Mokoena
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - H Steyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - A Hugo
- Department of Animal Science, University of the Free State, Bloemfontein, South Africa
| | - T Dix-Peek
- Department of Internal Medicine, University of Witwatersrand, Johannesburg, South Africa
| | - C Dickens
- Department of Internal Medicine, University of Witwatersrand, Johannesburg, South Africa
| | - O M N Gcilitshana
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - O Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - J Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - C H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa.
| |
Collapse
|
34
|
Fusco-Almeida AM, de Matos Silva S, dos Santos KS, de Lima Gualque MW, Vaso CO, Carvalho AR, Medina-Alarcón KP, Pires ACMDS, Belizario JA, de Souza Fernandes L, Moroz A, Martinez LR, Ruiz OH, González Á, Mendes-Giannini MJS. Alternative Non-Mammalian Animal and Cellular Methods for the Study of Host-Fungal Interactions. J Fungi (Basel) 2023; 9:943. [PMID: 37755051 PMCID: PMC10533014 DOI: 10.3390/jof9090943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
In the study of fungal pathogenesis, alternative methods have gained prominence due to recent global legislation restricting the use of mammalian animals in research. The principle of the 3 Rs (replacement, reduction, and refinement) is integrated into regulations and guidelines governing animal experimentation in nearly all countries. This principle advocates substituting vertebrate animals with other invertebrate organisms, embryos, microorganisms, or cell cultures. This review addresses host-fungus interactions by employing three-dimensional (3D) cultures, which offer more faithful replication of the in vivo environment, and by utilizing alternative animal models to replace traditional mammals. Among these alternative models, species like Caenorhabditis elegans and Danio rerio share approximately 75% of their genes with humans. Furthermore, models such as Galleria mellonella and Tenebrio molitor demonstrate similarities in their innate immune systems as well as anatomical and physiological barriers, resembling those found in mammalian organisms.
Collapse
Affiliation(s)
- Ana Marisa Fusco-Almeida
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Samanta de Matos Silva
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Kelvin Sousa dos Santos
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Marcos William de Lima Gualque
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Carolina Orlando Vaso
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Angélica Romão Carvalho
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Kaila Petrolina Medina-Alarcón
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Ana Carolina Moreira da Silva Pires
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Jenyffie Araújo Belizario
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Lígia de Souza Fernandes
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Andrei Moroz
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Luis R. Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA
- Center for Immunology and Transplantation, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Orville Hernandez Ruiz
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
- Cellular and Molecular Biology Group University of Antioquia, Corporation for Biological Research, Medellin 050010, Colombia
| | - Ángel González
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Maria José Soares Mendes-Giannini
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| |
Collapse
|
35
|
Muthubharathi BC, Ravichandiran V, Balamurugan K. Distinct global metabolomic profiles of the model organism Caenorhabditis elegans during interactions with Staphylococcus aureus and Salmonella enterica Serovar Typhi. Mol Omics 2023; 19:574-584. [PMID: 37272185 DOI: 10.1039/d3mo00040k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The interactive network of hosts with pathogenic microbes is still questionable. It has been hypothesized and reported that the host shows altered regulatory mechanisms for different pathogens. Several studies using transcriptomics and proteomics revealed the altered pathways and sequential regulations displayed by the host during bacterial interactions. Still, there is a gap in understanding the triggering molecule at transcriptomic and proteomic levels due to the lack of the knowledge of the interactive metabolites produced during their interactions. In this study, the global metabolomic approach was performed in the nematode model organism Caenorhabditis elegans upon exposure to a Gram-negative bacteria, Salmonella enterica Serovar Typhi, and a Gram-positive bacteria, Staphylococcus aureus, and the whole metabolome was categorized as endo-metabolome (internally produced) and exo-metabolome (externally releasing). The extracted metabolites were subjected to liquid chromatography mass spectrometry (ESI-LC/qToF-MS/MS). In total 5578, 4554 and 4046 endo-metabolites and 4451, 3625 and 1281 exo-metabolites were identified in C. elegans when exposed to E. coli OP50, S. Typhi and S. aureus, respectively. Both the multivariate and univariate analyses were performed. The variation in endo- and exo-metabolome during candidate bacterial interactions was observed. The results indicated that, during S. aureus interaction, the exclusively enriched metabolites were significantly involved in alpha-linoleic acid metabolism. Similarly, the exclusively enriched metabolites during the interaction of S. Typhi were significantly involved in the phosphatidylinositol signalling system. The whole metabolomic profile presented here will build the scope to understand the role of metabolites and the respective pathways in host response during the early period of bacterial infections.
Collapse
|
36
|
Doering KRS, Ermakova G, Taubert S. Nuclear hormone receptor NHR-49 is an essential regulator of stress resilience and healthy aging in Caenorhabditis elegans. Front Physiol 2023; 14:1241591. [PMID: 37645565 PMCID: PMC10461480 DOI: 10.3389/fphys.2023.1241591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023] Open
Abstract
The genome of Caenorhabditis elegans encodes 284 nuclear hormone receptor, which perform diverse functions in development and physiology. One of the best characterized of these is NHR-49, related in sequence and function to mammalian hepatocyte nuclear factor 4α and peroxisome proliferator-activated receptor α. Initially identified as regulator of lipid metabolism, including fatty acid catabolism and desaturation, additional important roles for NHR-49 have since emerged. It is an essential contributor to longevity in several genetic and environmental contexts, and also plays vital roles in the resistance to several stresses and innate immune response to infection with various bacterial pathogens. Here, we review how NHR-49 is integrated into pertinent signaling circuits and how it achieves its diverse functions. We also highlight areas for future investigation including identification of regulatory inputs that drive NHR-49 activity and identification of tissue-specific gene regulatory outputs. We anticipate that future work on this protein will provide information that could be useful for developing strategies to age-associated declines in health and age-related human diseases.
Collapse
Affiliation(s)
- Kelsie R. S. Doering
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Glafira Ermakova
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
- Edwin S. H. Leong Centre for Healthy Aging, The University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Venkatesh SR, Gupta A, Singh V. Amphid sensory neurons of Caenorhabditis elegans orchestrate its survival from infection with broad classes of pathogens. Life Sci Alliance 2023; 6:e202301949. [PMID: 37258276 PMCID: PMC10233725 DOI: 10.26508/lsa.202301949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023] Open
Abstract
The survival of a host during infection relies on its ability to rapidly sense the invading pathogen and mount an appropriate response. The bacterivorous nematode Caenorhabditis elegans lacks most of the traditional pattern recognition mechanisms. In this study, we hypothesized that the 12 pairs of amphid sensory neurons in the heads of worms provide sensing capability and thus affect survival during infection. We tested animals lacking amphid neurons to three major classes of pathogens, namely-a Gram-negative bacterium Pseudomonas aeruginosa, a Gram-positive bacterium Enterococcus faecalis, and a pathogenic yeast Cryptococcus neoformans By using individual neuronal ablation lines or mutants lacking specific neurons, we demonstrate that some neurons broadly suppress the survival of the host and colonization of all pathogens, whereas other amphid neurons differentially regulate host survival during infection. We also show that the roles of some of these neurons are pathogen-specific, as seen with the AWB odor sensory neurons that promote survival only during infections with P aeruginosa Overall, our study reveals broad and specific roles for amphid neurons during infections.
Collapse
Affiliation(s)
- Siddharth R Venkatesh
- Department of Developmental Biology & Genetics, Indian Institute of Science, Bangalore, INDIA
| | - Anjali Gupta
- Center for Biosystems, Science and Engineering, Indian Institute of Science, Bangalore, INDIA
| | - Varsha Singh
- Department of Developmental Biology & Genetics, Indian Institute of Science, Bangalore, INDIA
- Center for Biosystems, Science and Engineering, Indian Institute of Science, Bangalore, INDIA
| |
Collapse
|
38
|
Rezaeianaran F, Gijs MAM. Difference in Intestine Content of Caenorhabditis elegans When Fed on Non-Pathogenic or Pathogenic Bacteria. MICROMACHINES 2023; 14:1386. [PMID: 37512697 PMCID: PMC10384281 DOI: 10.3390/mi14071386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
We investigated the bacterial food digestion and accumulation in wild-type adult Caenorhabditis elegans (C. elegans) worms that have fed on either non-pathogenic RFP-expressing Escherichia coli (E. coli) OP50 or pathogenic-RFP-expressing Pseudomonas aeruginosa (P. aeruginosa) PAO1 during the first 4 days of adulthood. Once the worms had completed their planned feeding cycles, they were loaded on microfluidic chips, where they were fixed to allow high-resolution z-stack fluorescence imaging of their intestines utilizing a Spinning Disk Confocal Microscope (SDCM) equipped with a high-resolution oil-immersion objective (60×). IMARIS software was used to visualize and analyze the obtained images, resulting in the production of three-dimensional constructs of the intestinal bacterial load. We discovered two distinct patterns for the bacteria-derived fluorescence signal in the intestine: (i) individual fluorescent spots, originating from intact bacteria, were present in the fluorescent E. coli-OP50-fed worms, and (ii) individual fluorescent spots (originating from intact bacteria) were dispersed in large regions of diffuse fluorescence (RDF), originating from disrupted bacteria, in fluorescent P. aeruginosa-PAO1-fed worms. We performed a semi-automated single-worm-resolution quantitative analysis of the intestinal bacterial load, which showed that the intestinal bacterial load generally increases with age of the worms, but more rapidly for the fluorescent P. aeruginosa-PAO1-fed worms.
Collapse
Affiliation(s)
- Farzad Rezaeianaran
- Laboratory of Microsystems, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Martin A M Gijs
- Laboratory of Microsystems, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
39
|
Lažetić V, Blanchard MJ, Bui T, Troemel ER. Multiple pals gene modules control a balance between immunity and development in Caenorhabditis elegans. PLoS Pathog 2023; 19:e1011120. [PMID: 37463170 PMCID: PMC10353827 DOI: 10.1371/journal.ppat.1011120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023] Open
Abstract
The immune system continually battles against pathogen-induced pressures, which often leads to the evolutionary expansion of immune gene families in a species-specific manner. For example, the pals gene family expanded to 39 members in the Caenorhabditis elegans genome, in comparison to a single mammalian pals ortholog. Our previous studies have revealed that two members of this family, pals-22 and pals-25, act as antagonistic paralogs to control the Intracellular Pathogen Response (IPR). The IPR is a protective transcriptional response, which is activated upon infection by two molecularly distinct natural intracellular pathogens of C. elegans-the Orsay virus and the fungus Nematocida parisii from the microsporidia phylum. In this study, we identify a previously uncharacterized member of the pals family, pals-17, as a newly described negative regulator of the IPR. pals-17 mutants show constitutive upregulation of IPR gene expression, increased immunity against intracellular pathogens, as well as impaired development and reproduction. We also find that two other previously uncharacterized pals genes, pals-20 and pals-16, are positive regulators of the IPR, acting downstream of pals-17. These positive regulators reverse the effects caused by the loss of pals-17 on IPR gene expression, immunity, and development. We show that the negative IPR regulator protein PALS-17 and the positive IPR regulator protein PALS-20 colocalize inside and at the apical side of intestinal epithelial cells, which are the sites of infection for IPR-inducing pathogens. In summary, our study demonstrates that several pals genes from the expanded pals gene family act as ON/OFF switch modules to regulate a balance between organismal development and immunity against natural intracellular pathogens in C. elegans.
Collapse
Affiliation(s)
- Vladimir Lažetić
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Michael J. Blanchard
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Theresa Bui
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
40
|
Angusamy A, Balasubramanian V, Arunmurugan B, Arunachalam K, Issac Abraham SVP, Murugesan S, Krishnasamy B, Sundaram J, Arumugam VR. Anti-infective potential of plant-derived quorum sensing inhibitors against multi-drug resistant human and aquatic bacterial pathogens. World J Microbiol Biotechnol 2023; 39:147. [PMID: 37022521 DOI: 10.1007/s11274-023-03578-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
The present study intended to decipher the anti-infective potential of bioactive phytocompounds, such as rosmarinic acid, morin, naringin, chlorogenic acid, and mangiferin, against aquatic and human bacterial pathogens using Artemia spp. nauplii and Caenorhabditis elegans as animal models, respectively. Initially, the test compounds were screened against the QS traits in Vibrio spp., such as bioluminescence production and biofilm formation. The test compounds effectively inhibited the bioluminescence in V. harveyi. Further, the confocal laser scanning microscopic analysis revealed that these natural compounds could efficiently reduce the clumping morphology, a characteristic biofilm formation in Vibrio spp., without inhibiting bacterial growth. The results of in vivo analysis showed a significant increase in the survival of Artemia spp. nauplii infected with Vibrio spp. upon exposure to these compounds. Moreover, the compounds used in this study were already proven and reported for their quorum sensing inhibitory efficacy against Pseudomonas aeruginosa. Hence, the anti-infective efficacy of these compounds against P. aeruginosa (PAO1) and its clinical isolates (AS1 and AS2) was studied using C. elegans as a live animal model system. The results of time-killing assay deciphered that rosmarinic acid and naringin are being the most effective ones in rescuing the animals from P. aeruginosa infection followed by morin, mangiferin, and chlorogenic acid. Further, the toxicity results revealed that these compounds did not show any lethal effect on C. elegans and Artemia spp. nauplii at the tested concentrations. In conclusion, the phytochemicals used in this study were effective in controlling the QS-regulated virulence traits in Vibrio spp. and P. aeruginosa infections in Artemia spp. nauplii and C. elegans animal model systems, respectively.
Collapse
Affiliation(s)
- Annapoorani Angusamy
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600 025, India
| | - Vigneshkumar Balasubramanian
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
- Conservation of Coastal and Marine Resources (CMR) division, National Centre for Sustainable Coastal Management (NCSCM), Anna University campus, Tamil Nadu, 600 025, Chennai, India
| | - Balaji Arunmurugan
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
| | - Kannapan Arunachalam
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
- Department of Food Science and Technology, School of Agricultural Science, Shanghai Jiao Tong University, Shanghai, China
| | - Sybiya Vasantha Packiavathy Issac Abraham
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore, Tamil Nadu, 641 114, India
| | - Sivaranjani Murugesan
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK, Canada
| | | | - Janarthanan Sundaram
- Department of Zoology, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600 025, India
| | - Veera Ravi Arumugam
- Department of Biotechnology, Alagappa University, Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
41
|
Cochran JP, Unrine JM, Coyne M, Tsyusko OV. Multiple stressor effects on a model soil nematode, Caenorhabditis elegans: Combined effects of the pathogen Klebsiella pneumoniae and zinc oxide nanoparticles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 865:161307. [PMID: 36596421 PMCID: PMC9896629 DOI: 10.1016/j.scitotenv.2022.161307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Research utilizing the model soil nematode Caenorhabditis elegans has revealed that agriculturally relevant nanoparticles (NP), such as zinc oxide NP (ZnONP), cause toxicity at low concentrations and disrupt molecular pathways of pathogen resistance. However, in most nanotoxicity assessments, model organisms are exposed to a single stressor but in nature organisms are affected by multiple sources of stress, including infections, which might exacerbate or mitigate negative effects of NP exposure. Thus, to expand our understanding of the environmental consequences of released NP, this project examined the synergistic/antagonistic effects of ZnONP on C. elegans infected with a common pathogen, Klebsiella pneumoniae. Individual exposures of C. elegans to ZnONP, zinc sulfate (Zn2+ ions) or K. pneumoniae significantly decreased nematode reproduction compared to controls. To assess the combined stress of ZnONP and K. pneumoniae, C. elegans were exposed to equitoxic EC30 concentrations of ZnONP (or Zn ions) and K. pneumoniae. After the combined exposure there was no decrease in reproduction. This complete elimination of reproductive toxicity was unexpected because exposures were conducted at EC30 Zn concentrations and reproductive toxicity due to Zn should have occurred. Amelioration of the pathogen effects by Zn are partially explained by the Zn impact on the K. pneumoniae biofilm. Quantitative assessments showed that external biofilm production and estimated colony forming units (CFU) of K. pneumoniae within the nematodes were significantly decreased. Taken together, our results suggest that during the combined exposure of C. elegans to both stressors Zn in ionic or particulate form inhibits K. pneumoniae ability to colonize nematode's intestine through decreasing pathogen biofilm formation. This highlights the unpredictable nature of combined stressor effects, calling into question the utility of exposures in simplified laboratory media.
Collapse
Affiliation(s)
- Jarad P Cochran
- Department of Plant and Soil Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Jason M Unrine
- Department of Plant and Soil Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA; Kentucky Water Resources Research Institute, University of Kentucky, Lexington, KY, USA
| | - Mark Coyne
- Department of Plant and Soil Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Olga V Tsyusko
- Department of Plant and Soil Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
42
|
Majumder S, Sackey T, Viau C, Park S, Xia J, Ronholm J, George S. Genomic and phenotypic profiling of Staphylococcus aureus isolates from bovine mastitis for antibiotic resistance and intestinal infectivity. BMC Microbiol 2023; 23:43. [PMID: 36803552 PMCID: PMC9940407 DOI: 10.1186/s12866-023-02785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is one of the prevalent etiological agents of contagious bovine mastitis, causing a significant economic burden on the global dairy industry. Given the emergence of antibiotic resistance (ABR) and possible zoonotic spillovers, S aureus from mastitic cattle pose threat to both veterinary and public health. Therefore, assessment of their ABR status and pathogenic translation in human infection models is crucial. RESULTS In this study, 43 S. aureus isolates associated with bovine mastitis obtained from four different Canadian provinces (Alberta, Ontario, Quebec, and Atlantic provinces) were tested for ABR and virulence through phenotypic and genotypic profiling. All 43 isolates exhibited crucial virulence characteristics such as hemolysis, and biofilm formation, and six isolates from ST151, ST352, and ST8 categories showed ABR. Genes associated with ABR (tetK, tetM, aac6', norA, norB, lmrS, blaR, blaZ, etc.), toxin production (hla, hlab, lukD, etc.), adherence (fmbA, fnbB, clfA, clfB, icaABCD, etc.), and host immune invasion (spa, sbi, cap, adsA, etc.) were identified by analyzing whole-genome sequences. Although none of the isolates possessed human adaptation genes, both groups of ABR and antibiotic-susceptible isolates demonstrated intracellular invasion, colonization, infection, and death of human intestinal epithelial cells (Caco-2), and Caenorhabditis elegans. Notably, the susceptibilities of S. aureus towards antibiotics such as streptomycin, kanamycin, and ampicillin were altered when the bacteria were internalized in Caco-2 cells and C. elegans. Meanwhile, tetracycline, chloramphenicol, and ceftiofur were comparatively more effective with ≤ 2.5 log10 reductions of intracellular S. aureus. CONCLUSIONS This study demonstrated the potential of S. aureus isolated from mastitis cows to possess virulence characteristics enabling invasion of intestinal cells thus calling for developing therapeutics capable of targeting drug-resistant intracellular pathogens for effective disease management.
Collapse
Affiliation(s)
- Satwik Majumder
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Trisha Sackey
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Charles Viau
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Soyoun Park
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jianguo Xia
- grid.14709.3b0000 0004 1936 8649Institute of Parasitology, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Jennifer Ronholm
- grid.14709.3b0000 0004 1936 8649Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada ,grid.14709.3b0000 0004 1936 8649Department of Animal Science, Macdonald Campus, McGill University, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9 Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, Macdonald Campus, McGill University, Macdonald-Stewart Building, Room-1039, 21, 111 Lakeshore Ste Anne de Bellevue, Quebec, H9X 3V9, Canada.
| |
Collapse
|
43
|
Goswamy D, Gonzalez X, Labed SA, Irazoqui JE. C. elegans orphan nuclear receptor NHR-42 represses innate immunity and promotes lipid loss downstream of HLH-30/TFEB. Front Immunol 2023; 14:1094145. [PMID: 36860863 PMCID: PMC9968933 DOI: 10.3389/fimmu.2023.1094145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
In recent years, transcription factors of the Microphthalmia-TFE (MiT) family, including TFEB and TFE3 in mammals and HLH-30 in Caenorhabditis elegans, have emerged as important regulators of innate immunity and inflammation in invertebrates and vertebrates. Despite great strides in knowledge, the mechanisms that mediate downstream actions of MiT transcription factors in the context of innate host defense remain poorly understood. Here, we report that HLH-30, which promotes lipid droplet mobilization and host defense, induces the expression of orphan nuclear receptor NHR-42 during infection with Staphylococcus aureus. Remarkably, NHR-42 loss of function promoted host infection resistance, genetically defining NHR-42 as an HLH-30-controlled negative regulator of innate immunity. During infection, NHR-42 was required for lipid droplet loss, suggesting that it is an important effector of HLH-30 in lipid immunometabolism. Moreover, transcriptional profiling of nhr-42 mutants revealed wholesale activation of an antimicrobial signature, of which abf-2, cnc-2, and lec-11 were important for the enhanced survival of infection of nhr-42 mutants. These results advance our knowledge of the mechanisms by which MiT transcription factors promote host defense, and by analogy suggest that TFEB and TFE3 may similarly promote host defense via NHR-42-homologous nuclear receptors in mammals.
Collapse
Affiliation(s)
| | | | | | - Javier E. Irazoqui
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
44
|
Heat-Killed Staphylococcus aureus Induces Bone Mass Loss through Telomere Erosion. Int J Mol Sci 2023; 24:ijms24043179. [PMID: 36834587 PMCID: PMC9960843 DOI: 10.3390/ijms24043179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
The mechanism of systemic osteoporosis caused by chronic infection is not completely clear, and there is a lack of reasonable interventions for this disease. In this study, heat-killed S. aureus (HKSA) was applied to simulate the inflammation caused by the typical clinical pathogen and to explore the mechanism of systemic bone loss caused by it. In this study, we found that the systemic application of HKSA caused bone loss in mice. Further exploration found that HKSA caused cellular senescence, telomere length shortening, and telomere dysfunction-induced foci (TIF) in limb bones. As a well-known telomerase activator, cycloastragenol (CAG) significantly alleviated HKSA-induced telomere erosion and bone loss. These results suggested that telomere erosion in bone marrow cells is a possible mechanism of HKSA-induced bone loss. CAG may protect against HKSA-induced bone loss by alleviating telomere erosion in bone marrow cells.
Collapse
|
45
|
Lažetić V, Blanchard MJ, Bui T, Troemel ER. Multiple pals gene modules control a balance between immunity and development in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524171. [PMID: 36711775 PMCID: PMC9882112 DOI: 10.1101/2023.01.15.524171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The immune system continually battles against pathogen-induced pressures, which often leads to the evolutionary expansion of immune gene families in a species-specific manner. For example, the pals gene family expanded to 39 members in the Caenorhabditis elegans genome, in comparison to a single mammalian pals ortholog. Our previous studies have revealed that two members of this family, pals-22 and pals-25 , act as antagonistic paralogs to control the Intracellular Pathogen Response (IPR). The IPR is a protective transcriptional response, which is activated upon infection by two molecularly distinct natural intracellular pathogens of C. elegans - the Orsay virus and the fungus Nematocida parisii from the microsporidia phylum. In this study, we identify a previously uncharacterized member of the pals family, pals-17 , as a newly described negative regulator of the IPR. pals-17 mutants show constitutive upregulation of IPR gene expression, increased immunity against intracellular pathogens, as well as impaired development and reproduction. We also find that two other previously uncharacterized pals genes, pals-20 and pals-16 , are positive regulators of the IPR, acting downstream of pals-17 . These positive regulators reverse the effects caused by the loss of pals-17 on IPR gene expression, immunity and development. We show that the negative IPR regulator protein PALS-17 and the positive IPR regulator protein PALS-20 colocalize inside intestinal epithelial cells, which are the sites of infection for IPR-inducing pathogens. In summary, our study demonstrates that several pals genes from the expanded pals gene family act as ON/OFF switch modules to regulate a balance between organismal development and immunity against natural intracellular pathogens in C. elegans . AUTHOR SUMMARY Immune responses to pathogens induce extensive rewiring of host physiology. In the short term, these changes are generally beneficial as they can promote resistance against infection. However, prolonged activation of immune responses can have serious negative consequences on host health, including impaired organismal development and fitness. Therefore, the balance between activating the immune system and promoting development must be precisely regulated. In this study, we used genetics to identify a gene in the roundworm Caenorhabditis elegans called pals-17 that acts as a repressor of the Intracellular Pathogen Response (IPR), a defense response against viral and microsporidian infections. We also found that pals-17 is required for the normal development of these animals. Furthermore, we identified two other pals genes, pals-20 and pals-16 , as suppressors of pals-17 mutant phenotypes. Finally, we found that PALS-17 and PALS-20 proteins colocalize inside intestinal cells, where viruses and microsporidia invade and replicate in the host. Taken together, our study demonstrates a balance between organismal development and immunity that is regulated by several genetic ON/OFF switch 'modules' in C. elegans .
Collapse
Affiliation(s)
- Vladimir Lažetić
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Michael J. Blanchard
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Theresa Bui
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States
| | - Emily R. Troemel
- School of Biological Sciences, University of California, San Diego, La Jolla, California, United States,Corresponding author
| |
Collapse
|
46
|
Karengera A, Bao C, Bovee TFH, Dinkla IJT, Murk AJ. A Multiplex Gene Expression Assay for Direct Measurement of RNA Transcripts in Crude Lysates of the Nematode Caenorhabditis elegans Used as a Bioanalytical Tool. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:130-142. [PMID: 36282018 PMCID: PMC10107722 DOI: 10.1002/etc.5505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/19/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Gene expression profiling in Caenorhabditis elegans has been demonstrated to be a potential bioanalytical tool to detect the toxic potency of environmental contaminants. The RNA transcripts of genes responding to toxic exposure can be used as biomarkers for detecting these toxins. For routine application in environmental quality monitoring, an easy-to-use multiplex assay is required to reliably quantify expression levels of these biomarkers. In the present study, a bead-based assay was developed to fingerprint gene expression in C. elegans by quantitating messenger RNAs (mRNAs) of multiple target genes directly from crude nematode lysates, circumventing RNA extraction and purification steps. The assay uses signal amplification rather than target amplification for direct measurement of toxin-induced RNA transcripts. Using a 50-gene panel, the expression changes of four candidate reference genes and 46 target mRNAs for various contaminants and wastewaters were successfully measured, and the expression profiles indicated the type of toxin present. Moreover, the multiplex assay response was in line with previous results obtained with more time-consuming reverse-transcription quantitative polymerase chain reaction and microarray analyses. In addition, the transcriptomic profiles of nematodes exposed to wastewater samples and extracts prepared from tissues of swimming crabs were evaluated. The profiles indicated the presence of organic pollutants. The present study illustrates the successful development of a multiplex fluorescent bead-based approach using nematode C. elegans crude lysates for gene expression profiling of target RNAs. This method can be used to routinely fingerprint the presence of toxic contaminants in environmental samples and to identify the most biologically active fraction of the contaminant mixture in a toxicity identification and evaluation approach. Environ Toxicol Chem 2023;42:130-142. © 2022 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Antoine Karengera
- Department of Animal Sciences, Marine Animal Ecology GroupWageningen UniversityWageningenThe Netherlands
- Wetsus, European Centre of Excellence for Sustainable Water TechnologyLeeuwardenThe Netherlands
| | - Cong Bao
- Department of Animal Sciences, Marine Animal Ecology GroupWageningen UniversityWageningenThe Netherlands
- Department of Analysis and Testing CenterYangtze Delta Region Institute of Tsinghua UniversityJiaxingChina
| | - Toine F. H. Bovee
- Wageningen Food Safety Research, Team Bioassays & BiosensorsWageningen University & ResearchWageningenThe Netherlands
| | - Inez J. T. Dinkla
- Wetsus, European Centre of Excellence for Sustainable Water TechnologyLeeuwardenThe Netherlands
| | - Albertinka J. Murk
- Department of Animal Sciences, Marine Animal Ecology GroupWageningen UniversityWageningenThe Netherlands
| |
Collapse
|
47
|
Mangu JCK, Stylianou M, Olsson PE, Jass J. Per- and polyfluoroalkyl substances enhance Staphylococcus aureus pathogenicity and impair host immune response. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 314:120294. [PMID: 36181932 DOI: 10.1016/j.envpol.2022.120294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/18/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
Per- and Poly-fluoroalkyl substances (PFAS) are major persistent environmental contaminants. Epidemiological studies have linked PFAS exposures to altered immunity and increased occurrence of infections in children. However, the mechanisms leading to immune susceptibility to bacterial infections remains unclear. To elucidate the mechanism, transcriptional alteration in the Caenorhabditis elegans model caused by a PFAS contaminated environmental water and two reconstituted PFAS solutions were evaluated using RNA-sequencing. PFAS affected the expression of several genes involved in C. elegans immune surveillance to Gram-positive bacteria (cpr-2, tag-38, spp-1, spp-5, clec-7, clec-172). The combined exposure to PFAS and Staphylococcus aureus significantly reduced C. elegans survival and increased intestinal membrane permeability. Furthermore, the growth of S. aureus in the presence of PFAS increased the expression of virulence genes, specifically, the virulence gene regulator saeR and α-hemolysin, hla, which resulted in increased hemolytic activity. The present study demonstrated that PFAS exposure not only increased C. elegans susceptibility to pathogens by reducing host immunity and increasing intestinal membrane permeability, but also increased bacteria virulence. This presents a broader implication for humans and other animals, where environmental contaminants simultaneously reduce host resilience, while, increasing microbial pathogenicity.
Collapse
Affiliation(s)
| | - Marios Stylianou
- The Life Science Centre-Biology, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Per-Erik Olsson
- The Life Science Centre-Biology, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Jana Jass
- The Life Science Centre-Biology, School of Science and Technology, Örebro University, Örebro, Sweden.
| |
Collapse
|
48
|
Ramesh P, Bajire SK, Kanichery A, Najar MA, Shastry RP, Prasad TSK. 6-Methylcoumarin rescues bacterial quorum sensing induced ribosome-inactivating stress in Caenorhabditis elegans. Microb Pathog 2022; 173:105833. [PMID: 36265737 DOI: 10.1016/j.micpath.2022.105833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Bacterial pathogenicity has for long posed severe effects on patient care. Pseudomonas aeruginosa is a common cause of hospital-acquired infections and nosocomial illnesses. It is known to infect the host by colonizing through quorum sensing and the production of exotoxins. METHODS The current effort is an analysis of proteomic alterations caused by P. aeruginosa PAO1 to study the effects of quorum sensing inhibitor 6-Methylcoumarin on PAO1 infectivity in the Caenorhabditis elegans model. RESULTS Through tandem mass tag-based quantitative proteomics approaches, 229 proteins were found to be differentially regulated in infection and upon inhibition. Among these, 34 proteins were found to be dysregulated in both infection and quorum-sensing inhibition conditions. Along with the dysregulation of proteins involved in host-pathogen interaction, PAO1 was found to induce ribosome-inactivating stress accompanied by the downregulating mitochondrial proteins. This in turn caused dysregulation of apoptosis. The expression of multiple proteins involved in ribosome biogenesis and structure, oxidative phosphorylation, and mitochondrial enzymes were altered due to infection. This mechanism, adapted by PAO1 to survive in the host, was inhibited by 6-Methylcoumarin by rescuing the downregulation of ribosomal and mitochondrial proteins. CONCLUSIONS Taken together, the data reflect the molecular alterations due to quorum sensing and the usefulness of inhibitors in controlling pathogenesis.
Collapse
Affiliation(s)
- Poornima Ramesh
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| | - Sukesh Kumar Bajire
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| | - Anagha Kanichery
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| | - Rajesh P Shastry
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, India.
| |
Collapse
|
49
|
Liudkovska V, Krawczyk PS, Brouze A, Gumińska N, Wegierski T, Cysewski D, Mackiewicz Z, Ewbank JJ, Drabikowski K, Mroczek S, Dziembowski A. TENT5 cytoplasmic noncanonical poly(A) polymerases regulate the innate immune response in animals. SCIENCE ADVANCES 2022; 8:eadd9468. [PMID: 36383655 PMCID: PMC9668313 DOI: 10.1126/sciadv.add9468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Innate immunity is the first line of host defense against pathogens. Here, through global transcriptome and proteome analyses, we uncover that newly described cytoplasmic poly(A) polymerase TENT-5 (terminal nucleotidyltransferase 5) enhances the expression of secreted innate immunity effector proteins in Caenorhabditis elegans. Direct RNA sequencing revealed that multiple mRNAs with signal peptide-encoding sequences have shorter poly(A) tails in tent-5-deficient worms. Those mRNAs are translated at the endoplasmic reticulum where a fraction of TENT-5 is present, implying that they represent its direct substrates. Loss of tent-5 makes worms more susceptible to bacterial infection. Notably, the role of TENT-5 in innate immunity is evolutionarily conserved. Its orthologs, TENT5A and TENT5C, are expressed in macrophages and induced during their activation. Analysis of macrophages devoid of TENT5A/C revealed their role in the regulation of secreted proteins involved in defense response. In summary, our study reveals cytoplasmic polyadenylation to be a previously unknown component of the posttranscriptional regulation of innate immunity in animals.
Collapse
Affiliation(s)
- Vladyslava Liudkovska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
- Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Paweł S Krawczyk
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Aleksandra Brouze
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
- Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Natalia Gumińska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
| | - Tomasz Wegierski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
| | - Dominik Cysewski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Zuzanna Mackiewicz
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
| | - Jonathan J Ewbank
- Aix Marseille University, INSERM, CNRS, CIML, Turing Centre for Living Systems, Marseille, France
| | - Krzysztof Drabikowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Seweryn Mroczek
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
- Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Andrzej Dziembowski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Trojdena 4, 02-109 Warsaw, Poland
- Faculty of Biology, University of Warsaw, Pawinskiego 5a, 02-106 Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Poland
| |
Collapse
|
50
|
Ren J, Sang Y, Aballay A. Cholinergic receptor-Wnt pathway controls immune activation by sensing intestinal dysfunction. Cell Rep 2022; 41:111575. [DOI: 10.1016/j.celrep.2022.111575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 09/09/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
|