1
|
Kaletsky R, Moore RS, Sengupta T, Seto R, Ceballos-Llera B, Murphy CT. Molecular requirements for C. elegans transgenerational epigenetic inheritance of pathogen avoidance. eLife 2025; 14:RP105673. [PMID: 40372780 PMCID: PMC12080996 DOI: 10.7554/elife.105673] [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] [Indexed: 05/16/2025] Open
Abstract
Bacteria are Caenorhabditis elegans' food, and worms are naturally attracted to many bacteria, including pathogenic Pseudomonas, preferring PA14 over laboratory Escherichia coli (OP50). Despite this natural attraction to PA14, prior PA14 exposure causes the worms to instead avoid PA14. This behavioral switch can happen quickly - even within the duration of the choice assay. We show that accurate assessment of the animals' true first choice requires the use of a paralytic (azide) to trap the worms at their initial choice, preventing the switch from attraction to avoidance of PA14 within the assay period. We previously discovered that exposure of C. elegans to 25°C plate-grown PA14 at 20°C for 24 hr not only leads to PA14 avoidance, but also to four generations of naïve progeny avoiding PA14, while other PA14 paradigms only cause P0 and/or F1 avoidance. We also showed that the transgenerational (P0-F4) epigenetic avoidance is mediated by P11, a small RNA produced by PA14. P11 is both necessary and sufficient for TEI of learned avoidance. P11 is highly expressed in our standard growth conditions (25°C on surfaces), but not in other conditions, suggesting that the reported failure to observe F2-F4 avoidance is likely due to the absence of P11 expression in PA14 in the experimenters' growth conditions. Additionally, we tested ~35 genes for involvement in TEI of learned pathogen avoidance. The conservation of multiple components of this sRNA TEI mechanism across C. elegans strains and in multiple Pseudomonas species suggests that this TEI behavior is likely to be physiologically important in wild conditions.
Collapse
Affiliation(s)
- Rachel Kaletsky
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| | - Rebecca S Moore
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
- Chronobiology and Sleep Institute, Department of NeurosciencePhiladelphiaUnited States
- Howard Hughes Medical Institute, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Titas Sengupta
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| | - Renee Seto
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| | - Borja Ceballos-Llera
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| |
Collapse
|
2
|
Gao D, Shen L, Lin Y, Huang S, Xie Z. Ferric Uptake Regulator Contributes to Pseudomonas donghuensis HYS-Induced Iron Metabolic Disruption in Caenorhabditis elegans. Microorganisms 2025; 13:1081. [PMID: 40431254 PMCID: PMC12114261 DOI: 10.3390/microorganisms13051081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Iron is essential for vital biological processes, with its metabolism closely linked to host-pathogen interactions. Pseudomonas donghuensis HYS, with its superior iron uptake capacity, demonstrates pronounced virulence toward Caenorhabditis elegans. However, the virulence mechanisms remain unexplored. Ferric uptake regulator (Fur) regulates iron homeostasis and pathogenicity in bacteria, yet its role in HYS-mediated C. elegans pathogenesis requires systematic investigation. In this study, comparing the pathogenic processes of HYS and P. aeruginosa PA14 revealed that HYS causes stronger, irreversible toxicity via distinct mechanisms. Transcriptomics revealed that HYS infection disrupts C. elegans iron metabolism pathways, specifically iron transport, and iron-sulfur cluster utilization. Fur was identified as a pivotal regulator in HYS virulence and was indispensable for its colonization. Specifically, Fur was critical for disrupting nematode iron metabolism, as fur deletion eliminated this effect. While Fur regulated two HYS siderophores, neither of them mediated in the iron metabolism disruption of C. elegans. Screening identified Fur-regulated virulence factors to further investigate the function of Fur in HYS virulence, particularly alkaline proteases, and type II secretion system components. This study highlight that HYS can disrupt the iron metabolism pathway in C. elegans; Fur serves as a pivotal positive regulator in HYS-induced damage, particularly in disrupting iron metabolism through a siderophore-independent pathway. These findings expand the understanding of Pseudomonas pathogenicity and Fur-mediated virulence regulation.
Collapse
Affiliation(s)
| | | | | | | | - Zhixiong Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China; (D.G.); (L.S.); (Y.L.); (S.H.)
| |
Collapse
|
3
|
Xiao Y, Xiang W, Gao D, Zheng B, Wang Z, Rong D, Bayram H, Ghiladi RA, Lorimer GH, Xie Z, Wang J. hmuSTUV operon positively regulates the alginate gene cluster to mediate the pathogenicity of Pseudomonas donghuensis HYS. Int J Biol Macromol 2025; 306:141430. [PMID: 40010467 DOI: 10.1016/j.ijbiomac.2025.141430] [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: 11/19/2024] [Revised: 01/27/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
Pseudomonas donghuensis HYS is highly virulent to Caenorhabditis elegans, but with mechanistic details that are not fully understood. The hmuSTUV operon was reported to participate in the synthesis of heme in Pseudomonas. However, the exact role of the hmuSTUV operon in Pseudomonas virulence has not been elucidated. In this study, we report for the first time that the hmuSTUV operon in P. donghuensis HYS causes host virulence, and that hmuS was a key gene for the toxicity of this operon. Furthermore, RNA-seq data showed that hmuS deletion inhibited alginate gene expression, thereby inhibiting biofilm formation. The hmuSTUV operon and alginate gene cluster are conserved in Pseudomonas. By constructing mutant strains carrying GFP, we found that the hmuS deletion reduced colonisation of HYS to the host gut. Moreover, the expression of the alginate gene cluster was controlled by the construction of a L-arabinose-inducible promoter. hmuS positively regulated alginate gene cluster expression, mediating bacterial virulence against C. elegans. In addition, HYS originating from the East Lake of Wuhan City was more pathogenic to zebrafish than any other pathogenic Pseudomonas, through impairment of zebrafish neurodevelopment and locomotor ability, by colonizing to the zebrafish brain. In conclusion, the hmuSTUV operon positively regulated the alg gene cluster, thereby disabling bacterial biofilm formation and colonisation to mediate bacterial pathogenicity to the host. These novel findings revealed the critical interaction between the hmuSTUV operon and the alg gene cluster in the bacterial virulence of Pseudomonas, providing new insights into Pseudomonas pathogenicity.
Collapse
Affiliation(s)
- Yaqian Xiao
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China; Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wang Xiang
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
| | - Donghao Gao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Bowen Zheng
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
| | - Zhiqian Wang
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
| | - Dechang Rong
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Department of Pulmonary Medicine, School of Medicine, Koc University, Istanbul, Turkey
| | - Reza A Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - George H Lorimer
- Department of Chemistry, University of Maryland, College Park, MD, USA
| | - Zhixiong Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Jun Wang
- Hubei Key Laboratory of Industry Microbiology, International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China.
| |
Collapse
|
4
|
Tse-Kang S, Wani KA, Pukkila-Worley R. Patterns of pathogenesis in innate immunity: insights from C. elegans. Nat Rev Immunol 2025:10.1038/s41577-025-01167-0. [PMID: 40247006 DOI: 10.1038/s41577-025-01167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
The cells in barrier tissues can distinguish pathogenic from commensal bacteria and target inflammatory responses only in the context of infection. As such, these cells must be able to identify pathogen infection specifically and not just the presence of an infectious organism, because many innocuous bacteria express the ligands that activate innate immunity in other contexts. Unravelling the mechanisms that underly this specificity, however, is challenging. Free-living nematodes, such as Caenorhabditis elegans, are faced with a similar dilemma, as they live in microorganism-rich habitats and eat bacteria as their source of nutrition. Nematodes lost canonical mechanisms of pattern recognition during their evolution and have instead evolved mechanisms to identify specific ligands or symptoms in the host that indicate active infection with an infectious microorganism. Here we review how C. elegans surveys for these patterns of pathogenesis to activate innate immune defences. Collectively, this work demonstrates that using C. elegans as an experimental platform to study host-pathogen interactions at barrier surfaces reveals primordial and fundamentally important principles of innate immune sensing in the animal branch of the tree of life.
Collapse
Affiliation(s)
- Samantha Tse-Kang
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Khursheed A Wani
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Read Pukkila-Worley
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
5
|
Khazaei M, Parsasefat M, Bahar A, Tahmasebi H, Oksenych V. Behavioral Cooperation or Conflict of Human Intestinal Roundworms and Microbiomes: A Bio-Activity Perspective. Cells 2025; 14:556. [PMID: 40214509 PMCID: PMC11988915 DOI: 10.3390/cells14070556] [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: 03/07/2025] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Human infections are greatly impacted by intestinal nematodes. These nematodes, which encompass the large roundworms, have a direct impact on human health and well-being due to their close cohabitation with the host's microorganisms. When nematodes infect a host, the microbiome composition changes, and this can impact the host's ability to control the parasites. We aimed to find out if the small intestinal roundworms produce substances that have antimicrobial properties and respond to their microbial environment, and if the immune and regulatory reactions to nematodes are altered in humans lacking gut microbes. There is no doubt that different nematodes living in the intestines can alter the balance of intestinal bacteria. Nonetheless, our knowledge about the parasite's influence on the gut microbiome remains restricted. The last two decades of study have revealed that the type of iron utilized can influence the activation of unique virulence factors. However, some roundworm proteins like P43, which makes up a large portion of the worm's excretory-secretory product, have an unknown role. This review explores how the bacterial iron regulatory network contributes to the adaptability of this opportunistic pathogen, allowing it to successfully infect nematodes in different host environments.
Collapse
Affiliation(s)
- Meisam Khazaei
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
| | - Malihe Parsasefat
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
| | - Aisa Bahar
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
- Biochemistry Department, Faculty of Medicine, Iran University of Medical Sciences, Tehran 14496-1453, Iran
| | - Hamed Tahmasebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
| | | |
Collapse
|
6
|
Flavell SW, Oren-Suissa M, Stern S. Sources of behavioral variability in C. elegans: Sex differences, individuality, and internal states. Curr Opin Neurobiol 2025; 91:102984. [PMID: 39986247 PMCID: PMC12038806 DOI: 10.1016/j.conb.2025.102984] [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: 05/22/2024] [Revised: 01/04/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
Animal behavior varies across different timescales. This includes rapid shifts in behavior as animals transition between states and long-term changes that develop throughout an organism's life. This review presents the contributions of sex differences, individuality, and internal states to behavioral variability in the roundworm Caenorhabditis elegans. Sex is determined by chromosome composition, which directs neuronal development through gene regulation and experience to shape dimorphic behaviors. Genetically identical individuals within the same sex and reared in the same conditions still display distinctive, long-lasting behavioral traits that are controlled by neuromodulatory systems. At all life stages, internal states within the individual, shaped by external factors like food and stress, modulate behavior over minutes to hours. The interplay between these factors gives rise to rich behavioral diversity in C. elegans. These factors impact behavior in a sequential manner, as genetic sex, individuality, and internal states influence behavior over progressively finer timescales.
Collapse
Affiliation(s)
- Steven W Flavell
- Howard Hughes Medical Institute, Picower Institute for Learning and Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Meital Oren-Suissa
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Shay Stern
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
7
|
Kilmury SLN, Graham KJ, Lamers RP, MacNeil LT, Burrows LL. Hyperpiliation, not loss of pilus retraction, reduces Pseudomonas aeruginosa pathogenicity. Microbiol Spectr 2025; 13:e0255824. [PMID: 39998244 PMCID: PMC11960060 DOI: 10.1128/spectrum.02558-24] [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: 10/18/2024] [Accepted: 01/25/2025] [Indexed: 02/26/2025] Open
Abstract
Type IVa pili (T4aP) are important virulence factors for many bacterial pathogens. Previous studies suggested that the retraction ATPase, PilT, modulates pathogenicity due to its critical role in pilus dynamics and twitching motility. Here we use a Caenorhabditis elegans slow-killing model to show that hyperpiliation, not loss of pilus retraction, reduces virulence of Pseudomonas aeruginosa strains PAK and PA14. Hyperactivating point mutations in the P. aeruginosa PilSR two-component system that controls transcription of the major pilin gene, pilA, increased levels of surface pili to the same extent as deleting pilT, without impairing twitching motility. These functionally hyperpiliated PilSR mutants had significant defects in pathogenicity that were rescued by deleting pilA or through disruption of hyperpiliation via deletion of the type III secretion system needle-length regulator, PscP. Hyperpiliated pilT deletion or pilO point mutants showed similar PilA-dependent impairments in virulence, validating the phenotype. Together, our data support a model where a surfeit of pili reduces virulence, potentially through the prevention of effective engagement of contact-dependent virulence factors. These findings suggest that the role of T4aP retraction in virulence should be revised.IMPORTANCEPseudomonas aeruginosa is a major contributor to hospital-acquired infections and particularly problematic due to its intrinsic resistance to many front-line antibiotics. Strategies to combat this and other important pathogens include the development of anti-virulence therapeutics. We show that the pathogenicity of P. aeruginosa is impaired when the amount of T4aP expressed on the cell surface increases, independent of the bacteria's ability to twitch. We propose that having excess T4aP on the cell surface may physically interfere with productive engagement of the contact-dependent type III secretion toxin delivery system. A better understanding of how T4aP modulate interaction of bacteria with target cells will improve the design of therapeutics targeting components involved in the regulation of T4aP expression and function to reduce the clinical burden of P. aeruginosa and other T4aP-expressing bacteria.
Collapse
Affiliation(s)
- Sara L. N. Kilmury
- Department of Biochemistry and Biomedical Sciences and the Michael G DGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Katherine J. Graham
- Department of Biochemistry and Biomedical Sciences and the Michael G DGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Ryan P. Lamers
- Department of Biochemistry and Biomedical Sciences and the Michael G DGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lesley T. MacNeil
- Department of Biochemistry and Biomedical Sciences and the Michael G DGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences and the Michael G DGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
8
|
Gainey DP, Shubin AV, Hunter CP. Reported transgenerational responses to Pseudomonas aeruginosa in Caenorhabditis elegans are not robust. eLife 2025; 13:RP100254. [PMID: 40135732 PMCID: PMC11942167 DOI: 10.7554/elife.100254] [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] [Indexed: 03/27/2025] Open
Abstract
We report our attempt to replicate reports of transgenerational epigenetic inheritance in Caenorhabditis elegans. Multiple laboratories report that C. elegans adults and their F1 embryos exposed to the pathogen Pseudomonas aeruginosa show pathogen aversion behavior and increased daf-7/TGFβ reporter gene expression. However, results from one group show persistence of both through the F4 generation. We failed to consistently detect either the avoidance response or elevated daf-7 expression beyond the F1 generation. We confirmed that the dsRNA transport proteins SID-1 and SID-2 are required for intergenerational (F1) inheritance of pathogen avoidance, but not for the F1 inheritance of elevated daf-7 expression. Reanalysis of RNA seq data provides additional evidence that this intergenerational inherited PA14 response may be mediated by small RNAs. The experimental methods are well-described, the source materials are readily available, including samples from the reporting laboratory, and we explored a variety of environmental conditions likely to account for lab-to-lab variability. None of these adjustments altered our results. We conclude that this example of transgenerational inheritance lacks robustness, confirm that the intergenerational avoidance response, but not the elevated daf-7p::gfp expression in F1 progeny, requires sid-1 and sid-2, and identify candidate siRNAs and target genes that may mediate this intergenerational response.
Collapse
Affiliation(s)
- Daniel Patrick Gainey
- Department of Molecular and Cellular Biology, Harvard University, Divinity Avenue, The Biological LaboratoryCambridgeUnited States
| | - Andrey V Shubin
- Department of Molecular and Cellular Biology, Harvard University, Divinity Avenue, The Biological LaboratoryCambridgeUnited States
| | - Craig P Hunter
- Department of Molecular and Cellular Biology, Harvard University, Divinity Avenue, The Biological LaboratoryCambridgeUnited States
| |
Collapse
|
9
|
Pitkänen M, Monteuuis G, Jackson CB, Matilainen O. Neopterin extends C. elegans lifespan in an ATFS-1-dependent manner. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001543. [PMID: 40191444 PMCID: PMC11971658 DOI: 10.17912/micropub.biology.001543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/09/2025]
Abstract
Neopterin, a byproduct of tetrahydrobiopterin synthesis, is commonly used as a biomarker for immune system activation. In addition to its role in immune responses, neopterin levels are known to increase with age. Its impact on longevity, however, remains unclear. Here, we demonstrate that neopterin supplementation extends lifespan in Caenorhabditis elegans . Additionally, neopterin shows moderate activation of the mitochondrial unfolded protein response (UPR mt ), and that the neopterin-mediated lifespan extension is dependent on ATFS-1 , the primary transcription factor regulating UPR mt . These findings highlight the need for further investigation into the biological functions and health-promoting effects of neopterin.
Collapse
Affiliation(s)
- Miina Pitkänen
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Geoffray Monteuuis
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Christopher B. Jackson
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Olli Matilainen
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Uusimaa, Finland
| |
Collapse
|
10
|
Yang C, Yao L, Chen D, Chen C, Li W, Tong H, Cheng Z, Yan Y, Lin L, Zhang J, Shi A. Endosomal catabolism of phosphatidylinositol 4,5-bisphosphate is fundamental in building resilience against pathogens. Protein Cell 2025; 16:161-187. [PMID: 39087719 PMCID: PMC11891140 DOI: 10.1093/procel/pwae041] [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: 01/31/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
Endosomes are characterized by the presence of various phosphoinositides that are essential for defining the membrane properties. However, the interplay between endosomal phosphoinositides metabolism and innate immunity is yet to be fully understood. Here, our findings highlight the evolutionary continuity of RAB-10/Rab10's involvement in regulating innate immunity. Upon infection of Caenorhabditis elegans with Pseudomonas aeruginosa, an increase in RAB-10 activity was observed in the intestine. Conversely, when RAB-10 was absent, the intestinal diacylglycerols (DAGs) decreased, and the animal's response to the pathogen was impaired. Further research revealed that UNC-16/JIP3 acts as an RAB-10 effector, facilitating the recruitment of phospholipase EGL-8 to endosomes. This leads to a decrease in endosomal phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and an elevation of DAGs, as well as the activation of the PMK-1/p38 MAPK innate immune pathway. It is noteworthy that the dimerization of UNC-16 is a prerequisite for its interaction with RAB-10(GTP) and the recruitment of EGL-8. Moreover, we ascertained that the rise in RAB-10 activity, due to infection, was attributed to the augmented expression of LET-413/Erbin, and the nuclear receptor NHR-25/NR5A1/2 was determined to be indispensable for this increase. Hence, this study illuminates the significance of endosomal PI(4,5)P2 catabolism in boosting innate immunity and outlines an NHR-25-mediated mechanism for pathogen detection in intestinal epithelia.
Collapse
Affiliation(s)
- Chao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Longfeng Yao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Changling Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hua Tong
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zihang Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanling Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anbing Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan 430030, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
11
|
Moses D, Needham CA, DeBardeleben H. Quiescent behavior in response to bacterial infection in C. elegans. MICROPUBLICATION BIOLOGY 2025; 2025. [PMID: 40114851 PMCID: PMC11923603 DOI: 10.17912/micropub.biology.001412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/17/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Sickness behaviors serve an important role in recovery from infection. Using the WorMotel and a motivated displacement assay, we show that C. elegans engages in quiescent behavior following infection with Serratia marcesens , a bacterial pathogen. This quiescence is increased with increasing severity of the infection. Furthermore, we show this behavior is distinct from stress-induced sleep due to a lack of feeding quiescence and regulation by sleep-inducing neurons.
Collapse
Affiliation(s)
- Daniel Moses
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania, United States
| | - Carly A Needham
- Biology and Health Sciences, Commonwealth University - Bloomsburg
| | | |
Collapse
|
12
|
Boujnane M, Zommiti M, Lesouhaitier O, Ferchichi M, Tahrioui A, Boukerb AM, Connil N. Pediococcus pentosaceus MZF16 Probiotic Strain Prevents In Vitro Cytotoxic Effects of Pseudomonas aeruginosa H103 and Prolongs the Lifespan of Caenorhabditis elegans. Pathogens 2025; 14:244. [PMID: 40137729 PMCID: PMC11945076 DOI: 10.3390/pathogens14030244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogenic bacterium, responsible for several life-threatening infections due to its multiple virulence factors and problematic multi-drug resistance, hence the necessity to find alternatives such as competitive probiotics. Pediococcus pentosaceus MZF16 is an LAB strain, isolated from traditional dried meat "Ossban", with high probiotic potential. Our study investigated the capacity of P. pentosaceus MZF16 to counteract P. aeruginosa H103 using several tests on intestinal cells (analysis of cytotoxicity, inflammation, adhesion/invasion) and on the in vivo Caenorhabditis elegans model. The effect of MZF16 on the quorum sensing of the pathogen was also examined. We found that P. pentosaceus MZF16 was able to reduce H103 cytotoxicity and inflammatory activity and prevented pathogen colonization and translocation across Caco-2/TC7 cells. MZF16 also exerted an anti-virulence effect by attenuating quorum-sensing (QS) molecules and pyoverdine production and extended C. elegans lifespan. The obtained results highlight the potential of P. pentosaceus MZF16 probiotic strain as an anti-Pseudomonas aeruginosa alternative and establish a basis for elucidating the mechanisms of P. pentosaceus MZF16 involved in countering P. aeruginosa virulence.
Collapse
Affiliation(s)
- Meryem Boujnane
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Mohamed Zommiti
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Olivier Lesouhaitier
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Mounir Ferchichi
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia;
| | - Ali Tahrioui
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Amine M. Boukerb
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| | - Nathalie Connil
- CBSA UR 4312, Université de Rouen Normandie, Université de Caen Normandie, Normandie Université, F-76000 Rouen, France; (M.B.); (M.Z.); (O.L.); (A.T.); (A.M.B.)
| |
Collapse
|
13
|
Afridi MI, Tu H. The Roles of Distinct Transcriptional Factors in the Innate Immunity of C. elegans. Cells 2025; 14:327. [PMID: 40072056 PMCID: PMC11899719 DOI: 10.3390/cells14050327] [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: 12/04/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Deleterious molecules or factors produced by pathogens can hinder the normal physiological functioning of organisms. In response to these survival challenges, organisms rely on innate immune signaling as their first line of defense, which regulates immune-responsive genes and antimicrobial peptides to protect against pathogenic infections. These genes are under the control of transcription factors, which are known to regulate the transcriptional activity of genes after binding to their regulatory sequences. Previous studies have employed Caenorhabditis elegans as a host-pathogen interaction model to demonstrate the essential role of different transcription factors in the innate immunity of worms. In this review, we summarize the advances made regarding the functioning of distinct transcription factors in the innate immune response upon pathogen infection. Finally, we discuss the open questions in the field, whose resolutions have the potential to expand our understanding of the mechanisms underlying the innate immunity of organisms.
Collapse
Affiliation(s)
- Muhammad Irfan Afridi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha 410082, China;
| | - Haijun Tu
- Shenzhen Research Institute, Hunan University, Shenzhen 518000, China
| |
Collapse
|
14
|
Banerjee S, Smalley NE, Saenjamsai P, Fehr A, Dandekar AA, Cabeen MT, Chandler JR. Quorum sensing regulation by the nitrogen phosphotransferase system in Pseudomonas aeruginosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636002. [PMID: 39975224 PMCID: PMC11838483 DOI: 10.1101/2025.02.01.636002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In the opportunistic pathogen Pseudomonas aeruginosa, the nitrogen-related phosphotransferase system (PTSNtr) influences multiple virulence behaviors. The PTSNtr is comprised of three enzymes: first PtsP, then the PtsO phosphocarrier, and the final PtsN phosphoacceptor. We previously showed that ptsP inactivation increases LasI-LasR quorum sensing, a system by which P. aeruginosa regulates genes in response to population density. LasI synthesizes a diffusible autoinducer that binds and activates the LasR receptor, which activates a feedback loop by increasing lasI expression. In this study, we examined the impact of the PTSNtr on quorum sensing. Disruption of ptsP increased the expression of some, but not all, tested quorum-controlled genes, including lasI, phzM (pyocyanin biosynthesis), hcnA (hydrogen cyanide biosynthesis), and, to a lesser extent, rsaL (quorum sensing regulator). Expression of these genes remained dependent on LasR and the autoinducer, whether provided endogenously or exogenously. Increased lasI expression in ΔptsP (or ΔptsO) cells was partly due to the presence of unphosphorylated PtsN, which alone was sufficient to elevate lasI expression. However, we observed residual increases in ΔptsP or ΔptsO cells even in the absence of PtsN, suggesting that PtsP and PtsO can regulate gene expression independent of PtsN. Indeed, genetically disrupting the PtsO phosphorylation site impacted gene expression in the absence of PtsN, and transcriptomic evidence suggested that PtsO and PtsN have distinct regulons. Our results expand our view of how the PTSNtr components function both within and apart from the classic phosphorylation cascade to regulate key virulence behaviors in P. aeruginosa.
Collapse
Affiliation(s)
- Samalee Banerjee
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | | | | | - Anthony Fehr
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | - Ajai A. Dandekar
- Department of Microbiology, University of Washington, Seattle, WA
| | - Matthew T. Cabeen
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma
| | | |
Collapse
|
15
|
Wang X, Liu C, Zhao M, Zhang K, Di Z, Liu H. An Artificial Neural Network for Image Classification Inspired by the Aversive Olfactory Learning Neural Circuit in Caenorhabditis elegans. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410637. [PMID: 39679791 PMCID: PMC11831476 DOI: 10.1002/advs.202410637] [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: 09/02/2024] [Revised: 11/11/2024] [Indexed: 12/17/2024]
Abstract
This study introduces an artificial neural network (ANN) for image classification task, inspired by the aversive olfactory learning neural circuit in Caenorhabditis elegans (C. elegans). Although artificial neural networks (ANNs) have demonstrated remarkable performance in various tasks, they still encounter challenges including excessive parameterization, high training costs and limited generalization capabilities, etc. C. elegans, boasting a simple nervous system consisting of merely 302 neurons, is capable of exhibiting complex behaviors such as aversive olfactory learning. This research pinpoints key neural circuit related to aversive olfactory learning in C. elegans by means of behavioral experiment and high-throughput RNA sequencing, and then translates it into an architecture of ANN for image classification. Furthermore, other ANNs for image classification with different architectures are constructed for comparative performance analysis to underscore the advantages of the bio-inspired designed architecture. The results show that the ANN inspired by the aversive olfactory learning neural circuit in C. elegans attains higher accuracy, greater consistency and faster convergence rate in the image classification task, particularly when dealing with more complex classification challenges. This study not only demonstrates the potential of bio-inspired design in improving the capabilities of ANNs but also offers a novel perspective and methodology for future ANNs design.
Collapse
Affiliation(s)
- Xuebin Wang
- Department of Systems ScienceFaculty of Arts and SciencesBeijing Normal UniversityZhuhai519087China
- International Academic Center of Complex SystemsBeijing Normal UniversityZhuhai519087China
- School of Systems ScienceBeijing Normal UniversityBeijing100875China
| | - Chunxiuzi Liu
- Department of Systems ScienceFaculty of Arts and SciencesBeijing Normal UniversityZhuhai519087China
- International Academic Center of Complex SystemsBeijing Normal UniversityZhuhai519087China
- School of Systems ScienceBeijing Normal UniversityBeijing100875China
| | - Meng Zhao
- School of Computer Science and EngineeringTianjin University of TechnologyTianjin300384China
| | - Ke Zhang
- Department of Systems ScienceFaculty of Arts and SciencesBeijing Normal UniversityZhuhai519087China
- International Academic Center of Complex SystemsBeijing Normal UniversityZhuhai519087China
| | - Zengru Di
- Department of Systems ScienceFaculty of Arts and SciencesBeijing Normal UniversityZhuhai519087China
- International Academic Center of Complex SystemsBeijing Normal UniversityZhuhai519087China
- School of Systems ScienceBeijing Normal UniversityBeijing100875China
| | - He Liu
- Department of Systems ScienceFaculty of Arts and SciencesBeijing Normal UniversityZhuhai519087China
- International Academic Center of Complex SystemsBeijing Normal UniversityZhuhai519087China
| |
Collapse
|
16
|
Chang TT, Chang CH, Hsiu-Chuan Liao V. Early life long-term exposure to aflatoxin B1 induces aging and alters innate immunity associated with SKN-1/Nrf2 in Caenorhabditis elegans. Chem Biol Interact 2025; 406:111349. [PMID: 39675545 DOI: 10.1016/j.cbi.2024.111349] [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: 08/15/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Aflatoxin B1 (AFB1), a known human carcinogen, represents the most toxic aflatoxin metabolite. Exposure to AFB1 causes increased oxidative stress and immunotoxicity, which are important factors contributing to aging. However, the role of AFB1-induced toxicity in altered innate immunity and aging remains largely unclear. The nematode Caenorhabditis elegans is a suitable model organism for studying aging and toxicology due to its well-studied molecular mechanisms and short life cycle. Effects of AFB1 at 1, 2.5, and 5 μM (312, 781, and 1561 μg/L) on growth, reproduction, and lifespan were examined. The Pseudomonas aeruginosa PA14 slow-killing assay was performed to investigate innate immunity, followed by studying the possible mechanisms using transgenic strains and qPCR analysis. The results showed that early life long-term AFB1 exposure (2.5 and 5 μM) delayed development, reduced reproduction, and shortened lifespan in C. elegans. Furthermore, in aged worms, AFB1 exposure caused a dose-dependent decrease in survival of C. elegans against P. aeruginosa PA14 infection. At adulthood day 4 in the presence of live Escherichia coli OP50, AFB1 (2.5 μM) significantly increased lipofuscin levels (a hallmark of aging) compared to adult day 0, whereas no increase in lipofuscin was observed in nematodes (adulthood day 4) fed with dead E. coli OP50. Additionally, the increased lipofuscin was abolished in the skn-1 mutant with either live or dead E. coli OP50. Furthermore, AFB1 suppressed intestinal SKN-1::GFP translocation. Two-way ANOVA analysis revealed that the activity of E. coli OP50 and AFB1 interactively affected the expression of genes: skn-1, gst-4, hsp-16.1, hsp-16.49, and hsp-70. Our findings highlight the role of AFB1-induced toxicity in altered innate immunity and aging through the involvement of the transcription factor SKN-1/Nrf2.
Collapse
Affiliation(s)
- Tzu-Ting Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 106, Taiwan
| | - Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 106, Taiwan.
| |
Collapse
|
17
|
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
|
18
|
Liu H, Chen P, Yang X, Hao F, Tian G, Shan Z, Qi B. Probiotics-sensing mechanism in neurons that initiates gut mitochondrial surveillance for pathogen defense. Cell Rep 2024; 43:115021. [PMID: 39602305 DOI: 10.1016/j.celrep.2024.115021] [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: 05/18/2023] [Revised: 08/24/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Animals constantly face microbial challenges, and microbe-mediated infection protection is crucial for host survival. Identifying specific bacteria and their interactions with host intracellular surveillance systems is important but challenging. Here, we develop a "probiotics" screening system that identifies Escherichia coli mutants, such as ΔymcB, which protect hosts from Pseudomonas aeruginosa PA14 infection by activating the mitochondrial unfolded protein response (UPRmt). Genetic screening reveals that MDSS-1, a neuronal transmembrane protein, is crucial for sensing ΔymcB and triggering intestinal UPRmt. MDSS-1 functions as a potential receptor in ASE neurons, detecting ΔymcB and transmitting signals through neuropeptides, GPCRs, Wnt signaling, and endopeptidase inhibitors to activate intestinal UPRmt and enhance protection. Constitutive activation of MDSS-1 in ASE neurons is sufficient to induce UPRmt and confer infection resistance. This study uncovers a neuron-intestine communication mechanism, where ASE neurons detect bacteria and modulate the intestinal mitochondrial surveillance system for host adaptation to pathogens.
Collapse
Affiliation(s)
- Huimin Liu
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Panpan Chen
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Xubo Yang
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - FanRui Hao
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Guojing Tian
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China
| | - Zhao Shan
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China.
| | - Bin Qi
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming, China.
| |
Collapse
|
19
|
Coakley AJ, Hruby AJ, Wang J, Bong A, Nair T, Ramos CM, Alcala A, Hicks D, Averbukh M, Dutta N, Moaddeli D, Siebrand C, Rogers MDLR, Sahay A, Curran SP, Mullen PJ, Benayoun BA, Garcia G, Higuchi-Sanabria R. Distinct mechanisms of non-autonomous UPR ER mediated by GABAergic, glutamatergic, and octopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.595950. [PMID: 38854121 PMCID: PMC11160609 DOI: 10.1101/2024.05.27.595950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The capacity to deal with stress declines during the aging process, and preservation of cellular stress responses is critical to healthy aging. The unfolded protein response of the endoplasmic reticulum (UPRER) is one such conserved mechanism, which is critical for the maintenance of several major functions of the ER during stress, including protein folding and lipid metabolism. Hyperactivation of the UPRER by overexpression of the major transcription factor, xbp-1s, solely in neurons drives lifespan extension as neurons send a neurotransmitter-based signal to other tissue to activate UPRER in a non-autonomous fashion. Previous work identified serotonergic, dopaminergic, and tyraminergic neurons in this signaling paradigm. To further expand our understanding of the neural circuitry that underlies the non-autonomous signaling of ER stress, we activated UPRER solely in glutamatergic, octopaminergic, and GABAergic neurons in C. elegans and paired whole-body transcriptomic analysis with functional assays. We found that UPRER-induced signals from glutamatergic neurons increased expression of canonical protein homeostasis pathways and octopaminergic neurons promoted pathogen response pathways; while minor, statistically significant changes were observed in lipid metabolism-related genes with GABAergic UPRER activation. These findings provide further evidence for the distinct role neuronal subtypes play in driving the diverse response to ER stress.
Collapse
Affiliation(s)
- Aeowynn J. Coakley
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Adam J. Hruby
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Jing Wang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Andrew Bong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Tripti Nair
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Carmen M. Ramos
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Athena Alcala
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Daniel Hicks
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Maxim Averbukh
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Naibedya Dutta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Darius Moaddeli
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Cynthia Siebrand
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
- Current: Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Mattias de los Rios Rogers
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095, United States
| | - Arushi Sahay
- Department of Cell & Molecular Biology, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sean P. Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Peter J. Mullen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | | | - Gilberto Garcia
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| | - Ryo Higuchi-Sanabria
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, United States
| |
Collapse
|
20
|
Caracciolo ME, Villela EV, Machado LDS, Barreto ML, Rosa ACDP, Lopes-Torres EJ. Nematode-bacteria interactions in bovine parasitic otitis. REVISTA BRASILEIRA DE PARASITOLOGIA VETERINARIA = BRAZILIAN JOURNAL OF VETERINARY PARASITOLOGY : ORGAO OFICIAL DO COLEGIO BRASILEIRO DE PARASITOLOGIA VETERINARIA 2024; 33:e019024. [PMID: 39774744 PMCID: PMC11756828 DOI: 10.1590/s1984-29612024081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 01/11/2025]
Abstract
Bovine parasitic otitis poses challenges in diagnosis, treatment and involves various agents, such as bacteria, fungi, mites, and nematodes. This study focused on the nematodes and bacteria isolated from the auditory canals of dairy cattle. A total of twenty samples were collected from dairy cattle in two states of Brazil. The results showed that Metarhabditis freitasi and M. costai nematodes were identified in 75% of samples. Bacterial species from the ear, identified via mass spectrometry, revealed that different strains were present in 65% of the cattle. Mycoplasma spp. were identified in 45% of samples through molecular techniques. Gram-negative bacteria and Mycoplasma spp. were exclusively found in nematode-infected cattle. Furthermore, the bacteria exhibited resistance to multiple antimicrobial classes, and demonstrating multiresistance. Electron microscopy revealed biofilm aggregates on the cuticle of Metarhabditis spp., suggesting a potential role of these nematodes in bacterial migration and interaction with nervous tissue. Thirteen bacterial strains demonstrated biofilm formation ability, indicating their potential pathogenic role. This research highlights the persistent and complex nature of parasitic otitis, emphasizing the significant role of nematode-bacteria associations in its pathogenicity. The presence of resistant strains and biofilm formation underscores the challenges in managing the diagnosis and treatment of bovine parasitic otitis.
Collapse
Affiliation(s)
- Makoto Enoki Caracciolo
- Laboratório de Helmintologia Romero Lascasas Porto, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro – UERJ, Rio de Janeiro, RJ, Brasil
- Centro Multiusuário para Análise de Fenômenos Biomédicos, Universidade do Estado do Amazonas – UEA, Manaus, AM, Brasil
| | - Erika Verissimo Villela
- Departamento de Microbiologia, Imunologia e Parasitologia. Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro – UERJ, Rio de Janeiro, RJ, Brasil
| | - Leandro dos Santos Machado
- Departamento de Saúde Coletiva Veterinária e Saúde Pública, Universidade Federal Fluminense – UFF, Niterói, RJ, Brasil
| | - Maria Lúcia Barreto
- Núcleo de Animais de Laboratório, Universidade Federal Fluminense – UFF, Niterói, RJ, Brasil
| | - Ana Cláudia de Paula Rosa
- Departamento de Microbiologia, Imunologia e Parasitologia. Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro – UERJ, Rio de Janeiro, RJ, Brasil
| | - Eduardo José Lopes-Torres
- Laboratório de Helmintologia Romero Lascasas Porto, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro – UERJ, Rio de Janeiro, RJ, Brasil
- Laboratório Multiusuário de Parasitologia, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro – UERJ, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
21
|
Xue F, Ragno M, Blackburn SA, Fasseas M, Maitra S, Liang M, Rai S, Mastroianni G, Tholozan F, Thompson R, Sellars L, Hall R, Saunter C, Weinkove D, Ezcurra M. New tools to monitor Pseudomonas aeruginosa infection and biofilms in vivo in C. elegans. Front Cell Infect Microbiol 2024; 14:1478881. [PMID: 39737329 PMCID: PMC11683784 DOI: 10.3389/fcimb.2024.1478881] [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: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Antimicrobial resistance is a growing health problem. Pseudomonas aeruginosa is a pathogen of major concern because of its multidrug resistance and global threat, especially in health-care settings. The pathogenesis and drug resistance of P. aeruginosa depends on its ability to form biofilms, making infections chronic and untreatable as the biofilm protects against antibiotics and host immunity. A major barrier to developing new antimicrobials is the lack of in vivo biofilm models. Standard microbiological testing is usually performed in vitro using planktonic bacteria, without representation of biofilms, reducing translatability. Here we develop tools to study both infection and biofilm formation by P. aeruginosa in vivo to accelerate development of strategies targeting infection and pathogenic biofilms. Methods Biofilms were quantified in vitro using Crystal Violet staining and fluorescence biofilm assays. For in vivo assays, C. elegans were infected with P. aeruginosa strains. Pathogenicity was quantified by measuring healthspan, survival and GFP fluorescence. Healthspan assays were performed using the WormGazerTM automated imaging technology. Results Using the nematode Caenorhabditis elegans and P. aeruginosa reporters combined with in vivo imaging we show that fluorescent P. aeruginosa reporters that form biofilms in vitro can be used to visualize tissue infection. Using automated tracking of C. elegans movement, we find that that the timing of this infection corresponds with a decline in health endpoints. In a mutant strain of P. aeruginosa lacking RhlR, a transcription factor that controls quorum sensing and biofilm formation, we find reduced capacity of P. aeruginosa to form biofilms, invade host tissues and negatively impact healthspan and survival. Discussion Our findings suggest that RhlR could be a new antimicrobial target to reduce P. aeruginosa biofilms and virulence in vivo and C. elegans could be used to more effectively screen for new drugs to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Feng Xue
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Martina Ragno
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | | | - Michael Fasseas
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - Sushmita Maitra
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - Mingzhi Liang
- School of Biosciences, University of Kent, Canterbury, United Kingdom
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Subash Rai
- The NanoVision Centre, Queen Mary University of London, London, United Kingdom
| | - Giulia Mastroianni
- The NanoVision Centre, Queen Mary University of London, London, United Kingdom
| | | | - Rachel Thompson
- Perfectus Biomed Group, Sci-Tech Daresbury, Chesire, United Kingdom
| | - Laura Sellars
- Perfectus Biomed Group, Sci-Tech Daresbury, Chesire, United Kingdom
| | - Rebecca Hall
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Chris Saunter
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
| | - David Weinkove
- Magnitude Biosciences Limited, NETPark Plexus, Sedgefield, United Kingdom
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Marina Ezcurra
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
22
|
St Ange J, Weng Y, Kaletsky R, Stevenson ME, Moore RS, Zhou S, Murphy CT. Adult single-nucleus neuronal transcriptomes of insulin signaling mutants reveal regulators of behavior and learning. CELL GENOMICS 2024; 4:100720. [PMID: 39637862 DOI: 10.1016/j.xgen.2024.100720] [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: 02/22/2024] [Revised: 09/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Gene expression in individual neurons can change during development to adulthood and can have large effects on behavior. Additionally, the insulin/insulin-like signaling (IIS) pathway regulates many of the adult functions of Caenorhabditis elegans, including learning and memory, via transcriptional changes. We used the deep resolution of single-nucleus RNA sequencing to define the adult transcriptome of each neuron in wild-type and daf-2 mutants, revealing expression differences between L4 larval and adult neurons in chemoreceptors, synaptic genes, and learning/memory genes. We used these data to identify adult new AWC-specific regulators of chemosensory function that emerge upon adulthood. daf-2 gene expression changes correlate with improved cognitive functions, particularly in the AWC sensory neuron that controls learning and associative memory; behavioral assays of AWC-specific daf-2 genes revealed their roles in cognitive function. Combining technology and functional validation, we identified conserved genes that function in specific adult neurons to control behavior, including learning and memory.
Collapse
Affiliation(s)
- 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
| | - Yifei Weng
- 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
| | - Morgan E Stevenson
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rebecca S Moore
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Shiyi Zhou
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; 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
|
23
|
Schlichter Kadosh Y, Muthuraman S, Nisaa K, Ben-Zvi A, Karsagi Byron DL, Shagan M, Brandis A, Mehlman T, Gopas J, Saravana Kumar R, Kushmaro A. Pseudomonas aeruginosa quorum sensing and biofilm attenuation by a di-hydroxy derivative of piperlongumine (PL-18). Biofilm 2024; 8:100215. [PMID: 39148892 PMCID: PMC11326495 DOI: 10.1016/j.bioflm.2024.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/01/2024] [Accepted: 07/13/2024] [Indexed: 08/17/2024] Open
Abstract
Bacterial communication, Quorum Sensing (QS), is a target against virulence and prevention of antibiotic-resistant infections. 16 derivatives of Piperlongumine (PL), an amide alkaloid from Piper longum L., were screened for QS inhibition. PL-18 had the best QSI activity. PL-18 inhibited the lasR-lasI, rhlR-rhlI, and pqs QS systems of Pseudomonas aeruginosa. PL-18 inhibited pyocyanin and rhamnolipids that are QS-controlled virulence elements. Iron is an essential element for pathogenicity, biofilm formation and resilience in harsh environments, its uptake was inhibited by PL-18. Pl-18 significantly reduced the biofilm biovolume including in established biofilms. PL-18-coated silicon tubes significantly inhibited biofilm formation. The transcriptome study of treated P. aeruginosa showed that PL-18 indeed reduced the expression of QS and iron homeostasis related genes, and up regulated sulfur metabolism related genes. Altogether, PL-18 inhibits QS, virulence, iron uptake, and biofilm formation. Thus, PL-18 should be further developed against bacterial infection, antibiotic resistance, and biofilm formation.
Collapse
Affiliation(s)
- Yael Schlichter Kadosh
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Khairun Nisaa
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anat Ben-Zvi
- Department of Life Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Danit Lisa Karsagi Byron
- Department of Civil and Environmental Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marilou Shagan
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Gopas
- Department of Microbiology, Immunology and Genetics Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | | | - Ariel Kushmaro
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva, Israel
- The Ilse Katz Center for Nanoscale Science and Technology, Ben Gurion University of the Negev, Beer Sheva, Israel
- School of Sustainability and Climate Change, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
24
|
Ghosh A, Singh J. Translation initiation or elongation inhibition triggers contrasting effects on Caenorhabditis elegans survival during pathogen infection. mBio 2024; 15:e0248524. [PMID: 39347574 PMCID: PMC11559039 DOI: 10.1128/mbio.02485-24] [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: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
Diverse microbial pathogens are known to attenuate host protein synthesis. Consequently, the host mounts a defense response against protein translation inhibition, leading to increased transcript levels of immune genes. The seemingly paradoxical upregulation of immune gene transcripts in response to blocked protein synthesis suggests that the defense mechanism against translation inhibition may not universally benefit host survival. However, a comprehensive assessment of host survival on pathogens upon blockage of different stages of protein synthesis is currently lacking. Here, we investigate the impact of knockdown of various translation initiation and elongation factors on the survival of Caenorhabditis elegans exposed to Pseudomonas aeruginosa. Intriguingly, we observe opposing effects on C. elegans survival depending on whether translation initiation or elongation is inhibited. While translation initiation inhibition enhances survival, elongation inhibition decreases it. Transcriptomic studies reveal that translation initiation inhibition activates a bZIP transcription factor ZIP-2-dependent innate immune response that protects C. elegans from P. aeruginosa infection. In contrast, inhibiting translation elongation triggers both ZIP-2-dependent and ZIP-2-independent immune responses that, while effective in clearing the infection, are detrimental to the host. Thus, our findings reveal the opposing roles of translation initiation and elongation inhibition in C. elegans survival during P. aeruginosa infection, highlighting distinct transcriptional reprogramming that may underlie these differences. IMPORTANCE Several microbial pathogens target host protein synthesis machinery, potentially limiting the innate immune responses of the host. In response, hosts trigger a defensive response, elevating immune gene transcripts. This counterintuitive response can have either beneficial or harmful effects on host survival. In this study, we conduct a comprehensive analysis of the impact of knocking down various translation initiation and elongation factors on the survival of Caenorhabditis elegans exposed to Pseudomonas aeruginosa. Intriguingly, inhibiting initiation and elongation factors has contrasting effects on C. elegans survival. Inhibiting translation initiation activates immune responses that protect the host from bacterial infection, while inhibiting translation elongation induces aberrant immune responses that, although clear the infection, are detrimental to the host. Our study reveals divergent roles of translation initiation and elongation inhibition in C. elegans survival during P. aeruginosa infection and identifies differential transcriptional reprogramming that could underlie these differences.
Collapse
Affiliation(s)
- Annesha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| | - Jogender Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab, India
| |
Collapse
|
25
|
Gabaldón C, Karakuzu O, Garsin DA. SKN-1 activation during infection of Caenorhabditis elegans requires CDC-48 and endoplasmic reticulum proteostasis. Genetics 2024; 228:iyae131. [PMID: 39166513 PMCID: PMC11538416 DOI: 10.1093/genetics/iyae131] [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: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
During challenge of Caenorhabditis elegans with human bacterial pathogens such as Pseudomonas aeruginosa and Enterococcus faecalis, the elicited host response can be damaging if not properly controlled. The activation of Nrf (nuclear factor erythroid-related factor)/CNC (Cap-n-collar) transcriptional regulators modulates the response by upregulating genes that neutralize damaging molecules and promote repair processes. Activation of the C. elegans Nrf ortholog, SKN-1, is tightly controlled by a myriad of regulatory mechanisms, but a central feature is an activating phosphorylation accomplished by the p38 mitogen-activated kinase (MAPK) cascade. In this work, loss of CDC-48, an AAA+ ATPase, was observed to severely compromise SKN-1 activation on pathogen and we sought to understand the mechanism. CDC-48 is part of the endoplasmic reticulum (ER)-associated degradation (ERAD) complex where it functions as a remodeling chaperone enabling the translocation of proteins from the ER to the cytoplasm for degradation by the proteosome. Interestingly, one of the proteins retrotranslocated by ERAD, a process necessary for its activation, is SKN-1A, the ER isoform of SKN-1. However, we discovered that SKN-1A is not activated by pathogen exposure in marked contrast to the cytoplasmic-associated isoform SKN-1C. Rather, loss of CDC-48 blocks the antioxidant response normally orchestrated by SKN-1C by strongly inducing the unfolded protein response (UPRER). The data are consistent with the model of these 2 pathways being mutually inhibitory and support the emerging paradigm in the field of coordinated cooperation between different stress responses.
Collapse
Affiliation(s)
- Carolaing Gabaldón
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ozgur Karakuzu
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Danielle A Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
26
|
Sun X, Xiao F, Su Y, Li Z, Yu X, Parales RE, Li L. Cyclic di-GMP incorporates the transcriptional factor FleQ03 in Pseudomonas syringae MB03 to elicit biofilm-dependent resistance in response to Caenorhabditis elegans predation. J Invertebr Pathol 2024; 207:108189. [PMID: 39251105 DOI: 10.1016/j.jip.2024.108189] [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: 03/23/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Bacteria usually form biofilms as a defense mechanism against predation by bacterivorous nematodes. In this context, the second messenger c-di-GMP from the wild-type Pseudomonas syringae MB03 actuates the transcriptional factor FleQ03 to elicit biofilm-dependent nematicidal activity against Caenorhabditis elegans N2. P. syringae MB03 cells exhibited nematicidal activity and c-di-GMP content in P. syringae MB03 cells was increased after feeding to nematodes. Expression of a diguanylate cyclase (DGC) gene in P. syringae MB03 resulted in an increased c-di-GMP content, biofilm yield and nematicidal activity, whereas converse effects were obtained when expressing a phosphodiesterase (PDE) gene. Molecular docking and isothermal titration calorimetry assays verified the affinity activity between c-di-GMP and the FleQ03 protein. The disruption of the fleQ03 gene in P. syringae MB03, while increasing c-di-GMP content, significantly diminished both biofilm formation and nematicidal activity. Interestingly, P. syringae MB03 formed a full-body biofilm around the worms against predation, probably extending from the tail to the head, whereas it was not observed in the fleQ03 gene disrupted cells. Thus, we hypothesized that c-di-GMP incorporated FleQ03 to reinforce bacterial biofilm and biofilm-dependent pathogenicity in response to C. elegans predation, providing insights into a possible means of resisting bacterivorous nematodes by bacteria in natural ecosystems.
Collapse
Affiliation(s)
- Xiaowen Sun
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Fan Xiao
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwei Su
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhi Li
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun Yu
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Rebecca E Parales
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, USA.
| | - Lin Li
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
27
|
Gainey DP, Shubin AV, Hunter CP. Reported transgenerational responses to Pseudomonas aeruginosa in C. elegans are not robust. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596941. [PMID: 39554072 PMCID: PMC11565720 DOI: 10.1101/2024.06.01.596941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Here we report our attempt to replicate reports of transgenerational epigenetic inheritance in Caenorhabditis elegans. Published results from multiple laboratories show that C. elegans adults and their F1 embryos exposed to the pathogen Pseudomonas aeruginosa show pathogen aversion behavior and a pathogen exposure-dependent increase in daf-7/TGFβ reporter gene expression. However, results from one group show persistence of the aversion behavior and elevated daf-7 expression in the F2, F3, and F4 generations. In contrast, we failed to consistently detect either the pathogen avoidance response or elevated daf-7 expression beyond the F1 generation. We did confirm that the dsRNA transport proteins SID-1 and SID-2 are required for the intergenerational (F1) inheritance of pathogen avoidance, but not for the F1 inheritance of elevated daf-7 expression. Furthermore, our reanalysis of RNA seq data provides additional evidence that this intergenerational inherited PA14 response may be mediated by small RNAs. The experimental methods are well-described, the source materials are readily available, including samples from the reporting laboratory, and we explored a variety of environmental conditions likely to account for lab-to-lab variability. None of these adjustments altered our results. We conclude that this example of transgenerational inheritance lacks robustness, confirm that the intergenerational avoidance response, but not the elevated daf-7p::gfp expression in F1 progeny, requires sid-1 and sid-2, and identify candidate siRNAs and target genes that may mediate this intergenerational response.
Collapse
Affiliation(s)
- D. Patrick Gainey
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, The Biological Laboratory, Cambridge, MA 02138
| | - Andrey V. Shubin
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, The Biological Laboratory, Cambridge, MA 02138
| | - Craig P. Hunter
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, The Biological Laboratory, Cambridge, MA 02138
| |
Collapse
|
28
|
Wang Z, Zhang Q, Jiang Y, Zhou J, Tian Y. ASI-RIM neuronal axis regulates systemic mitochondrial stress response via TGF-β signaling cascade. Nat Commun 2024; 15:8997. [PMID: 39426950 PMCID: PMC11490647 DOI: 10.1038/s41467-024-53093-9] [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: 02/16/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Morphogens play a critical role in coordinating stress adaptation and aging across tissues, yet their involvement in neuronal mitochondrial stress responses and systemic effects remains unclear. In this study, we reveal that the transforming growth factor beta (TGF-β) DAF-7 is pivotal in mediating the intestinal mitochondrial unfolded protein response (UPRmt) in Caenorhabditis elegans under neuronal mitochondrial stress. Two ASI sensory neurons produce DAF-7, which targets DAF-1/TGF-β receptors on RIM interneurons to orchestrate a systemic UPRmt response. Remarkably, inducing mitochondrial stress specifically in ASI neurons activates intestinal UPRmt, extends lifespan, enhances pathogen resistance, and reduces both brood size and body fat levels. Furthermore, dopamine positively regulates this UPRmt activation, while GABA acts as a systemic suppressor. This study uncovers the intricate mechanisms of systemic mitochondrial stress regulation, emphasizing the vital role of TGF-β in metabolic adaptations that are crucial for organismal fitness and aging during neuronal mitochondrial stress.
Collapse
Affiliation(s)
- Zihao Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100093, Beijing, China
| | - Qian Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Yayun Jiang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100093, Beijing, China
| | - Jun Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100093, Beijing, China
| | - Ye Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100093, Beijing, China.
| |
Collapse
|
29
|
Hallacy T, Yonar A, Ringstad N, Ramanathan S. Compressed sensing based approach identifies modular neural circuitry driving learned pathogen avoidance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588911. [PMID: 39464156 PMCID: PMC11507717 DOI: 10.1101/2024.04.10.588911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
An animal's survival hinges on its ability to integrate past information to modify future behavior. The nematode C. elegans adapts its behavior based on prior experiences with pathogen exposure, transitioning from attraction to avoidance of the pathogen. A systematic screen for the neural circuits that integrate the information of previous pathogen exposure to modify behavior has not been feasible because of the lack of tools for neuron type specific perturbations. We overcame this challenge using methods based on compressed sensing to efficiently determine the roles of individual neuron types in learned avoidance behavior. Our screen revealed that distinct sets of neurons drive exit from lawns of pathogenic bacteria and prevent lawn re-entry. Using calcium imaging of freely behaving animals and optogenetic perturbations, we determined the neural dynamics that regulate one key behavioral transition after infection: stalled re-entry into bacterial lawns. We find that key neuron types govern pathogen lawn specific stalling but allow the animal to enter nonpathogenic E. coli lawns. Our study shows that learned pathogen avoidance requires coordinated transitions in discrete neural circuits and reveals the modular structure of this complex adaptive behavioral response to infection.
Collapse
Affiliation(s)
| | - Abdullah Yonar
- Departments of Molecular and Cellular Biology, and of Stem Cell and Regenerative Biology, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Niels Ringstad
- Department of Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sharad Ramanathan
- Departments of Molecular and Cellular Biology, and of Stem Cell and Regenerative Biology, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
30
|
Tse-Kang SY, Wani KA, Peterson ND, Page A, Humphries F, Pukkila-Worley R. Intestinal immunity in C. elegans is activated by pathogen effector-triggered aggregation of the guard protein TIR-1 on lysosome-related organelles. Immunity 2024; 57:2280-2295.e6. [PMID: 39299238 PMCID: PMC11464196 DOI: 10.1016/j.immuni.2024.08.013] [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: 02/16/2024] [Revised: 06/19/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024]
Abstract
Toll/interleukin-1/resistance (TIR)-domain proteins with enzymatic activity are essential for immunity in plants, animals, and bacteria. However, it is not known how these proteins function in pathogen sensing in animals. We discovered that the lone enzymatic TIR-domain protein in the nematode C. elegans (TIR-1, homolog of mammalian sterile alpha and TIR motif-containing 1 [SARM1]) was strategically expressed on the membranes of a specific intracellular compartment called lysosome-related organelles. The positioning of TIR-1 on lysosome-related organelles enables intestinal epithelial cells in the nematode C. elegans to survey for pathogen effector-triggered host damage. A virulence effector secreted by the bacterial pathogen Pseudomonas aeruginosa alkalinized and condensed lysosome-related organelles. This pathogen-induced morphological change in lysosome-related organelles triggered TIR-1 multimerization, which engaged its intrinsic NAD+ hydrolase (NADase) activity to activate the p38 innate immune pathway and protect the host against microbial intoxication. Thus, TIR-1 is a guard protein in an effector-triggered immune response, which enables intestinal epithelial cells to survey for pathogen-induced host damage.
Collapse
Affiliation(s)
- Samantha Y Tse-Kang
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Khursheed A Wani
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Nicholas D Peterson
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Amanda Page
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Fiachra Humphries
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Read Pukkila-Worley
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
31
|
Di Bernardo M, León Guerrero VL, Sutoski JC, Hardy WR, MacNeil LT. SHC-3: a previously unidentified C. elegans Shc family member functions in the insulin-like signaling pathway to enhance survival during L1 arrest. Genetics 2024; 228:iyae093. [PMID: 38861412 PMCID: PMC11630764 DOI: 10.1093/genetics/iyae093] [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: 11/21/2023] [Revised: 11/21/2023] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
Shc (Src homologous and collagen) proteins function in many different signaling pathways where they mediate phosphorylation-dependent protein-protein interactions. These proteins are characterized by the presence of two phosphotyrosine-binding domains, an N-terminal PTB and a C-terminal SH2. We describe a previously unrecognized Caenorhabditis elegans Shc gene, shc-3 and characterize its role in stress response. Both shc-3 and shc-1 are required for long-term survival in L1 arrest and survival in heat stress, however, they do not act redundantly but rather play distinct roles in these processes. Loss of shc-3 did not further decrease survival of daf-16 mutants in L1 arrest, suggesting that like SHC-1, SHC-3 functions in the insulin-like signaling pathway. In the absence of SHC-3, DAF-16 nuclear entry and exit are slowed, suggesting that SHC-3 is required for rapid changes in DAF-16 signaling.
Collapse
Affiliation(s)
- Mercedes Di Bernardo
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
| | - Victoria L León Guerrero
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
| | - Jacob C Sutoski
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
| | - William Rod Hardy
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main St W, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
32
|
Tse-Kang SY, Pukkila-Worley R. Lysosome-related organelle integrity suppresses TIR-1 aggregation to restrain toxic propagation of p38 innate immunity. Cell Rep 2024; 43:114674. [PMID: 39299237 PMCID: PMC11492801 DOI: 10.1016/j.celrep.2024.114674] [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: 02/16/2024] [Revised: 06/19/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
Innate immunity in bacteria, plants, and animals requires the specialized subset of Toll/interleukin-1/resistance gene (TIR) domain proteins that are nicotinamide adenine dinucleotide (NAD+) hydrolases. Aggregation of these TIR proteins engages their enzymatic activity, but it is unknown how this protein multimerization is regulated. Here, we discover that TIR oligomerization is controlled to prevent immune toxicity. We find that p38 propagates its own activation in a positive feedback loop, which promotes the aggregation of the lone enzymatic TIR protein in the nematode C. elegans (TIR-1, homologous to human sterile alpha and TIR motif-containing 1 [SARM1]). We perform a forward genetic screen to determine how the p38 positive feedback loop is regulated. We discover that the integrity of the specific lysosomal subcompartment that expresses TIR-1 is actively maintained to limit inappropriate TIR-1 aggregation on the membranes of these organelles, which restrains toxic propagation of p38 innate immunity. Thus, innate immunity in C. elegans intestinal epithelial cells is regulated by specific control of TIR-1 multimerization.
Collapse
Affiliation(s)
- Samantha Y Tse-Kang
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Read Pukkila-Worley
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
33
|
Roberts CG, Kaur S, Ogden AJ, Divine ME, Warren GD, Kang D, Kirienko NV, Geurink PP, Mulder MP, Nakayasu ES, McDermott JE, Adkins JN, Aballay A, Pruneda JN. A functional screen for ubiquitin regulation identifies an E3 ligase secreted by Pseudomonas aeruginosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613774. [PMID: 39345563 PMCID: PMC11430079 DOI: 10.1101/2024.09.18.613774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Ubiquitin signaling controls many aspects of eukaryotic biology, including targeted protein degradation and immune defense. Remarkably, invading bacterial pathogens have adapted secreted effector proteins that hijack host ubiquitination to gain control over host responses. These ubiquitin-targeted effectors can exhibit, for example, E3 ligase or deubiquitinase activities, often without any sequence or structural homology to eukaryotic ubiquitin regulators. Such convergence in function poses a challenge to the discovery of additional bacterial virulence factors that target ubiquitin. To overcome this, we have developed a workflow to harvest natively secreted bacterial effectors and functionally screen them for ubiquitin regulatory activities. After benchmarking this approach on diverse ligase and deubiquitinase activities from Salmonella Typhimurium, Enteropathogenic Escherichia coli, and Shigella flexneri, we applied it to the identification of a cryptic E3 ligase activity secreted by Pseudomonas aeruginosa. We identified an unreported P. aeruginosa E3 ligase, which we have termed Pseudomonas Ub ligase 1 (PUL-1), that resembles none of the other E3 ligases previously established in or outside of the eukaryotic system. Importantly, in an animal model of P. aeruginosa infection, PUL-1 ligase activity plays an important role in regulating virulence. Thus, our workflow for the functional identification of ubiquitin-targeted effector proteins carries promise for expanding our appreciation of how host ubiquitin regulation contributes to bacterial pathogenesis.
Collapse
Affiliation(s)
- Cameron G. Roberts
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Supender Kaur
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aaron J. Ogden
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Michael E. Divine
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Gus D. Warren
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Donghoon Kang
- Department of Biosciences, Rice University, Houston, TX 77005, USA
| | | | - Paul P. Geurink
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique P.C. Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jason E. McDermott
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Joshua N. Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alejandro Aballay
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jonathan N. Pruneda
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
34
|
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
|
35
|
Abstract
Behavioral plasticity allows animals to modulate their behavior based on experience and environmental conditions. Caenorhabditis elegans exhibits experience-dependent changes in its behavioral responses to various modalities of sensory cues, including odorants, salts, temperature, and mechanical stimulations. Most of these forms of behavioral plasticity, such as adaptation, habituation, associative learning, and imprinting, are shared with other animals. The C. elegans nervous system is considerably tractable for experimental studies-its function can be characterized and manipulated with molecular genetic methods, its activity can be visualized and analyzed with imaging approaches, and the connectivity of its relatively small number of neurons are well described. Therefore, C. elegans provides an opportunity to study molecular, neuronal, and circuit mechanisms underlying behavioral plasticity that are either conserved in other animals or unique to this species. These findings reveal insights into how the nervous system interacts with the environmental cues to generate behavioral changes with adaptive values.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yuichi Iino
- Department of Biological Sciences, University of Tokyo, Tokyo 113-0032, Japan
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, UK
- Department of Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
36
|
Woods KE, Akhter S, Rodriguez B, Townsend KA, Smith N, Smith B, Wambua A, Craddock V, Abisado-Duque RG, Santa EE, Manson DE, Oakley BR, Hancock LE, Miao Y, Blackwell HE, Chandler JR. Characterization of natural product inhibitors of quorum sensing reveals competitive inhibition of Pseudomonas aeruginosa RhlR by ortho-vanillin. Microbiol Spectr 2024; 12:e0068124. [PMID: 39046261 PMCID: PMC11370260 DOI: 10.1128/spectrum.00681-24] [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: 03/14/2024] [Accepted: 05/15/2024] [Indexed: 07/25/2024] Open
Abstract
Quorum sensing (QS) is a cell-cell signaling system that enables bacteria to coordinate population density-dependent changes in behavior. This chemical communication pathway is mediated by diffusible N-acyl L-homoserine lactone signals and cytoplasmic signal-responsive LuxR-type receptors in Gram-negative bacteria. As many common pathogenic bacteria use QS to regulate virulence, there is significant interest in disrupting QS as a potential therapeutic strategy. Prior studies have implicated the natural products salicylic acid, cinnamaldehyde, and other related benzaldehyde derivatives as inhibitors of QS in the opportunistic pathogen Pseudomonas aeruginosa, yet we lack an understanding of the mechanisms by which these compounds function. Herein, we evaluate the activity of a set of benzaldehyde derivatives using heterologous reporters of the P. aeruginosa LasR and RhlR QS signal receptors. We find that most tested benzaldehyde derivatives can antagonize LasR or RhlR reporter activation at micromolar concentrations, although certain molecules also cause mild growth defects and nonspecific reporter antagonism. Notably, several compounds showed promising RhlR or LasR-specific inhibitory activities over a range of concentrations below that causing toxicity. ortho-Vanillin, a previously untested compound, was the most promising within this set. Competition experiments against the native ligands for LasR and RhlR revealed that ortho-vanillin can interact competitively with RhlR but not with LasR. Overall, these studies expand our understanding of benzaldehyde activities in the LasR and RhlR receptors and reveal potentially promising effects of ortho-vanillin as a small molecule QS modulator against RhlR. IMPORTANCE Quorum sensing (QS) regulates many aspects of bacterial pathogenesis and has attracted much interest as a target for anti-virulence therapies over the past 30 years, for example, antagonists of the LasR and RhlR QS receptors in Pseudomonas aeruginosa. Potent and selective QS inhibitors remain relatively scarce. However, natural products have provided a bounty of chemical scaffolds with anti-QS activities, but their molecular mechanisms are poorly characterized. The current study serves to fill this void by examining the activity of an important and wide-spread class of natural product QS modulators, benzaldehydes, and related derivatives, in LasR and RhlR. We demonstrate that ortho-vanillin can act as a competitive inhibitor of RhlR, a receptor that has emerged and may supplant LasR in certain settings as a target for P. aeruginosa QS control. The results and insights provided herein will advance the design of chemical tools to study QS with improved activities and selectivities.
Collapse
Affiliation(s)
- Kathryn E. Woods
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Sana Akhter
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, USA
| | - Blanca Rodriguez
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Kade A. Townsend
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Nathan Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Ben Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Alice Wambua
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Vaughn Craddock
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | | | - Emma E. Santa
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Daniel E. Manson
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Berl R. Oakley
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Lynn E. Hancock
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Yinglong Miao
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, USA
| | - Helen E. Blackwell
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
37
|
Majdi C, Meffre P, Benfodda Z. Recent advances in the development of bacterial response regulators inhibitors as antibacterial and/or antibiotic adjuvant agent: A new approach to combat bacterial resistance. Bioorg Chem 2024; 150:107606. [PMID: 38968903 DOI: 10.1016/j.bioorg.2024.107606] [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: 03/25/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
The number of new antibacterial agents currently being discovered is insufficient to combat bacterial resistance. It is extremely challenging to find new antibiotics and to introduce them to the pharmaceutical market. Therefore, special attention must be given to find new strategies to combat bacterial resistance and prevent bacteria from developing resistance. Two-component system is a transduction system and the most prevalent mechanism employed by bacteria to respond to environmental changes. This signaling system consists of a membrane sensor histidine kinase that perceives environmental stimuli and a response regulator which acts as a transcription factor. The approach consisting of developing response regulators inhibitors with antibacterial activity or antibiotic adjuvant activity is a novel approach that has never been previously reviewed. In this review we report for the first time, the importance of targeting response regulators and summarizing all existing studies carried out from 2008 until now on response regulators inhibitors as antibacterial agents or / and antibiotic adjuvants. Moreover, we describe the antibacterial activity and/or antibiotic adjuvants activity against the studied bacterial strains and the mechanism of different response regulator inhibitors when it's possible.
Collapse
|
38
|
Wu Y, Li G, Tang H. Antibiotics Trigger Host Innate Immune Response via Microbiota-Brain Communication in C. elegans. Int J Mol Sci 2024; 25:8866. [PMID: 39201552 PMCID: PMC11354627 DOI: 10.3390/ijms25168866] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Besides their direct bactericidal effect, antibiotics have also been suggested to stimulate the host immune response to defend against pathogens. However, it remains unclear whether any antibiotics may stimulate the host immune response by affecting bacterial activity. In this study, reasoning that genetic mutations inhibit bacterial activities and, thereby, may mimic the effects of antibiotics, we performed genome-wide screening and identified 77 E. coli genes whose inactivation induces C. elegans cyp-14A4, representing an innate immune and detoxification response. Further analyses reveal that this host immune response can clearly be induced through either inactivating the E. coli respiratory chain via the bacterial cyoB mutation or using the antibiotic Q203, which is able to enhance host survival when encountering the pathogen Pseudomonas aeruginosa. Mechanistically, the innate immune response triggered by both the cyoB mutation and Q203 is found to depend on the host brain response, as evidenced by their reliance on the host neural gene unc-13, which is required for neurotransmitter release in head neurons. Therefore, our findings elucidate the critical involvement of the microbiota-brain axis in modulating the host immune response, providing mechanistic insights into the role of antibiotics in triggering the host immune response and, thus, facilitating host defense against pathogens.
Collapse
Affiliation(s)
- Yangyang Wu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Guanqun Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Hongyun Tang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
39
|
Espejo LS, Freitas S, Hofschneider V, Chang L, Antenor A, Balsa J, Haskins A, DeNicola D, Dang H, Hamming S, Kelser D, Sutphin GL. SICKO: Systematic Imaging of Caenorhabditis Killing Organisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.17.529009. [PMID: 39149381 PMCID: PMC11326133 DOI: 10.1101/2023.02.17.529009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Caenorhabditis elegans are an important model system for research on host-microbe interaction. Their rapid life cycle, short lifespan, and transparent body structure allow simple quantification of microbial load and the influence of microbial exposure on host survival. C. elegans host-microbe interaction studies typically examine group survival and infection severity at fixed timepoints. Here we present an imaging pipeline, Systematic Imaging of Caenorhabditis Killing Organisms (SICKO), that allows longitudinal characterization of microbes colonizing isolated C. elegans, enabling dynamic tracking of tissue colonization and host survival in the same animals. Using SICKO, we show that Escherichia coli or Pseudomonas aeruginosa gut colonization dramatically shortens C. elegans lifespan and that immunodeficient animals lacking pmk-1 are more susceptible to colonization but display similar colony growth relative to wild type. SICKO opens new avenues for detailed research into bacterial pathogenesis, the benefits of probiotics, and the role of the microbiome in host health.
Collapse
Affiliation(s)
- Luis S. Espejo
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Samuel Freitas
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | | | - Leah Chang
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Angelo Antenor
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Jonah Balsa
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Anne Haskins
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Destiny DeNicola
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Hope Dang
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Sage Hamming
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - Delaney Kelser
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| | - George L. Sutphin
- Molecular & Cellular Biology, University of Arizona, Tucson, AZ, USA, 85721
| |
Collapse
|
40
|
Pitkänen M, Matilainen O. Milk Fat Globule Membrane-Containing Protein Powder Promotes Fitness in Caenorhabditis elegans. Nutrients 2024; 16:2290. [PMID: 39064733 PMCID: PMC11280102 DOI: 10.3390/nu16142290] [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: 06/24/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Milk-derived peptides and milk fat globule membrane (MFGM) have gained interest as health-promoting food ingredients. However, the mechanisms by which these nutraceuticals modulate the function of biological systems often remain unclear. We utilized Caenorhabditis elegans to elucidate how MFGM-containing protein powder (MProPow), previously used in a clinical trial, affect the physiology of this model organism. Our results demonstrate that MProPow does not affect lifespan but promotes the fitness of the animals. Surprisingly, gene expression analysis revealed that MProPow decreases the expression of genes functioning on innate immunity, which also translates into reduced survival on pathogenic bacteria. One of the innate immunity-associated genes showing reduced expression upon MProPow supplementation is cpr-3, the homolog of human cathepsin B. Interestingly, knockdown of cpr-3 enhances fitness, but not in MProPow-treated animals, suggesting that MProPow contributes to fitness by downregulating the expression of this gene. In summary, this research highlights the value of C. elegans in testing the biological activity of food supplements and nutraceuticals. Furthermore, this study should encourage investigations into whether milk-derived peptides and MFGM mediate their beneficial effects through the modulation of cathepsin B expression in humans.
Collapse
Affiliation(s)
| | - Olli Matilainen
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790 Helsinki, Finland;
| |
Collapse
|
41
|
Wibisono P, Liu Y, Roberts KP, Baluya D, Sun J. Neuronal GPCR NMUR-1 regulates energy homeostasis in response to pathogen infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602733. [PMID: 39026696 PMCID: PMC11257582 DOI: 10.1101/2024.07.09.602733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
A key question in current immunology is how the innate immune system generates high levels of specificity. Our previous study in Caenorhabditis elegans revealed that NMUR-1, a neuronal G protein-coupled receptor homologous to mammalian receptors for the neuropeptide neuromedin U (NMU), regulates distinct innate immune responses to different bacterial pathogens. Here, by using quantitative proteomics and functional assays, we discovered that NMUR-1 regulates F1FO ATP synthase and ATP production in response to pathogen infection, and that such regulation contributes to NMUR-1-mediated specificity of innate immunity. We further demonstrated that ATP biosynthesis and its contribution to defense is neurally controlled by the NMUR-1 ligand CAPA-1 and its expressing neurons ASG. These findings indicate that NMUR-1 neural signaling regulates the specificity of innate immunity by controlling energy homeostasis as part of defense against pathogens. Our study provides mechanistic insights into the emerging roles of NMU signaling in immunity across animal phyla.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
- Genomics Core, Washington State University, Spokane, WA, USA
| | - Kenneth P Roberts
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Dodge Baluya
- Tissue Imaging, Metabolomics and Proteomics Laboratory, Washington State University, Pullman, WA, USA
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
42
|
Vassallo BG, Scheidel N, Fischer SEJ, Kim DH. Bacteria are a major determinant of Orsay virus transmission and infection in Caenorhabditis elegans. eLife 2024; 12:RP92534. [PMID: 38990923 PMCID: PMC11239179 DOI: 10.7554/elife.92534] [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] [Indexed: 07/13/2024] Open
Abstract
The microbiota is a key determinant of the physiology and immunity of animal hosts. The factors governing the transmissibility of viruses between susceptible hosts are incompletely understood. Bacteria serve as food for Caenorhabditis elegans and represent an integral part of the natural environment of C. elegans. We determined the effects of bacteria isolated with C. elegans from its natural environment on the transmission of Orsay virus in C. elegans using quantitative virus transmission and host susceptibility assays. We observed that Ochrobactrum species promoted Orsay virus transmission, whereas Pseudomonas lurida MYb11 attenuated virus transmission relative to the standard laboratory bacterial food Escherichia coli OP50. We found that pathogenic Pseudomonas aeruginosa strains PA01 and PA14 further attenuated virus transmission. We determined that the amount of Orsay virus required to infect 50% of a C. elegans population on P. lurida MYb11 compared with Ochrobactrum vermis MYb71 was dramatically increased, over three orders of magnitude. Host susceptibility was attenuated even further in the presence of P. aeruginosa PA14. Genetic analysis of the determinants of P. aeruginosa required for attenuation of C. elegans susceptibility to Orsay virus infection revealed a role for regulators of quorum sensing. Our data suggest that distinct constituents of the C. elegans microbiota and potential pathogens can have widely divergent effects on Orsay virus transmission, such that associated bacteria can effectively determine host susceptibility versus resistance to viral infection. Our study provides quantitative evidence for a critical role for tripartite host-virus-bacteria interactions in determining the transmissibility of viruses among susceptible hosts.
Collapse
Affiliation(s)
- Brian G Vassallo
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Noemie Scheidel
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Sylvia E J Fischer
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Dennis H Kim
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
43
|
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
|
44
|
Liu J, Lei M, Wang M, Chen S, Tu H. Protocol for survival assay of Caenorhabditis elegans to Pseudomonas aeruginosa PA14 infection. STAR Protoc 2024; 5:103070. [PMID: 38768031 PMCID: PMC11111813 DOI: 10.1016/j.xpro.2024.103070] [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: 11/28/2023] [Revised: 02/07/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024] Open
Abstract
The nematode Caenorhabditis elegans is a powerful model organism for studying the molecular and cellular mechanisms of innate immunity governed by the intestine. Here, we present a protocol to perform C. elegans survival assays to infection by the bacterial pathogen Pseudomonas aeruginosa PA14. Specifically, we describe steps for preparing C. elegans strains and PA14 bacteria for survival assays. This protocol will assist researchers to study genes involved in intestinal innate immunity and gut defense against pathogen infection. For complete details on the use and execution of this protocol, please refer to Liu et al.1 and Zheng et al.2.
Collapse
Affiliation(s)
- Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China.
| | - Ming Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Mengqi Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Shu Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
45
|
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
|
46
|
Fu S, Song W, Han X, Chen L, Shen L. Veratryl Alcohol Attenuates the Virulence and Pathogenicity of Pseudomonas aeruginosa Mainly via Targeting las Quorum-Sensing System. Microorganisms 2024; 12:985. [PMID: 38792814 PMCID: PMC11123940 DOI: 10.3390/microorganisms12050985] [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: 04/19/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that usually causes chronic infections and even death in patients. The treatment of P. aeruginosa infection has become more challenging due to the prevalence of antibiotic resistance and the slow pace of new antibiotic development. Therefore, it is essential to explore non-antibiotic methods. A new strategy involves screening for drugs that target the quorum-sensing (QS) system. The QS system regulates the infection and drug resistance in P. aeruginosa. In this study, veratryl alcohol (VA) was found as an effective QS inhibitor (QSI). It effectively suppressed the expression of QS-related genes and the subsequent production of virulence factors under the control of QS including elastase, protease, pyocyanin and rhamnolipid at sub-inhibitory concentrations. In addition, motility activity and biofilm formation, which were correlated with the infection of P. aeruginosa, were also suppressed by VA. In vivo experiments demonstrated that VA could weaken the pathogenicity of P. aeruginosa in Chinese cabbage, Drosophila melanogaster, and Caenorhabditis elegans infection models. Molecular docking, combined with QS quintuple mutant infection analysis, identified that the mechanism of VA could target the LasR protein of the las system mainly. Moreover, VA increased the susceptibility of P. aeruginosa to conventional antibiotics of tobramycin, kanamycin and gentamicin. The results firstly demonstrate that VA is a promising QSI to treat infections caused by P. aeruginosa.
Collapse
Affiliation(s)
| | | | | | | | - Lixin Shen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi’an 710069, China; (S.F.); (W.S.); (X.H.); (L.C.)
| |
Collapse
|
47
|
Zelikman S, Dudkevich R, Korenfeld-Tzemach H, Shmidov E, Levi-Ferber M, Shoshani S, Ben-Aroya S, Henis-Korenblit S, Banin E. PemB, a type III secretion effector in Pseudomonas aeruginosa, affects Caenorhabditis elegans life span. Heliyon 2024; 10:e29751. [PMID: 38681583 PMCID: PMC11053225 DOI: 10.1016/j.heliyon.2024.e29751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Pseudomonas aeruginosa is one of the leading nosocomial opportunistic pathogens causing acute and chronic infections. Among its main virulent factors is the Type III secretion system (T3SS) which enhances disease severity by delivering effectors to the host in a highly regulated manner. Despite its importance for virulence, only six T3SS-dependent effectors have been discovered so far. Previously, we identified two new potential effectors using a machine-learning algorithm approach. Here we demonstrate that one of these effectors, PemB, is indeed virulent. Using a live Caenorhabditis elegans infection model, we demonstrate this effector damages the integrity of the intestine barrier leading to the death of the host. Implementing a high-throughput assay using Saccharomyces cerevisiae, we identified several candidate proteins that interact with PemB. One of them, EFT1, has an ortholog in C. elegans (eef-2) and is also an essential gene and a well-known target utilized by different pathogens to induce toxicity to the worm. Accordingly, we found that by silencing the eef-2 gene in C. elegans, PemB could no longer induce its toxic effect. The current study further uncovers the complex machinery assisting P. aeruginosa virulence and may provide novel insight how to manage infection associated with this hard-to-treat pathogen.
Collapse
Affiliation(s)
- Shira Zelikman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Reut Dudkevich
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Hadar Korenfeld-Tzemach
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Esther Shmidov
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Mor Levi-Ferber
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Sivan Shoshani
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Shay Ben-Aroya
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Sivan Henis-Korenblit
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| | - Ehud Banin
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
- The Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Max and Anna Webb, 5290002, Ramat Gan, Israel
| |
Collapse
|
48
|
Marquina-Solis J, Feng L, Vandewyer E, Beets I, Hawk J, Colón-Ramos DA, Yu J, Fox BW, Schroeder FC, Bargmann CI. Antagonism between neuropeptides and monoamines in a distributed circuit for pathogen avoidance. Cell Rep 2024; 43:114042. [PMID: 38573858 PMCID: PMC11063628 DOI: 10.1016/j.celrep.2024.114042] [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: 04/16/2022] [Revised: 02/06/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
Pathogenic infection elicits behaviors that promote recovery and survival of the host. After exposure to the pathogenic bacterium Pseudomonas aeruginosa PA14, the nematode Caenorhabditis elegans modifies its sensory preferences to avoid the pathogen. Here, we identify antagonistic neuromodulators that shape this acquired avoidance behavior. Using an unbiased cell-directed neuropeptide screen, we show that AVK neurons upregulate and release RF/RYamide FLP-1 neuropeptides during infection to drive pathogen avoidance. Manipulations that increase or decrease AVK activity accelerate or delay pathogen avoidance, respectively, implicating AVK in the dynamics of avoidance behavior. FLP-1 neuropeptides drive pathogen avoidance through the G protein-coupled receptor DMSR-7, as well as other receptors. DMSR-7 in turn acts in multiple neurons, including tyraminergic/octopaminergic neurons that receive convergent avoidance signals from the cytokine DAF-7/transforming growth factor β. Neuromodulators shape pathogen avoidance through multiple mechanisms and targets, in agreement with the distributed neuromodulatory connectome of C. elegans.
Collapse
Affiliation(s)
- Javier Marquina-Solis
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Likui Feng
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA
| | | | - Isabel Beets
- Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Josh Hawk
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Daniel A Colón-Ramos
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Departments of Neuroscience and of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA; Instituto de Neurobiología José del Castillo, Recinto de Ciencias Médicas, Universidad de Puerto Rico, San Juan, PR 00901, USA; Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
| | - Jingfang Yu
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Bennett W Fox
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Cornelia I Bargmann
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
49
|
Pan S, Underhill SAM, Hamm CW, Stover MA, Butler DR, Shults CA, Manjarrez JR, Cabeen MT. Glycerol metabolism impacts biofilm phenotypes and virulence in Pseudomonas aeruginosa via the Entner-Doudoroff pathway. mSphere 2024; 9:e0078623. [PMID: 38501832 PMCID: PMC11036800 DOI: 10.1128/msphere.00786-23] [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: 12/14/2023] [Accepted: 02/23/2024] [Indexed: 03/20/2024] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous bacterium and a notorious opportunistic pathogen that forms biofilm structures in response to many environmental cues. Biofilm formation includes attachment to surfaces and the production of the exopolysaccharide Pel, which is present in both the PAO1 and PA14 laboratory strains of P. aeruginosa. Biofilms help protect bacterial cells from host defenses and antibiotics and abet infection. The carbon source used by the cells also influences biofilm, but these effects have not been deeply studied. We show here that glycerol, which can be liberated from host surfactants during infection, encourages surface attachment and magnifies colony morphology differences. We find that glycerol kinase is important but not essential for glycerol utilization and relatively unimportant for biofilm behaviors. Among downstream enzymes predicted to take part in glycerol utilization, Edd stood out as being important for glycerol utilization and for enhanced biofilm phenotypes in the presence of glycerol. Thus, gluconeogenesis and catabolism of anabolically produced glucose appear to impact not only the utilization of glycerol but also glycerol-stimulated biofilm phenotypes. Finally, waxworm moth larvae and nematode infection models reveal that interruption of the Entner-Doudoroff pathway, but not abrogation of glycerol phosphorylation, unexpectedly increases P. aeruginosa lethality in both acute and chronic infections, even while stimulating a stronger immune response by Caenorhabditis elegans.IMPORTANCEPseudomonas aeruginosa, the ubiquitous environmental bacterium and human pathogen, forms multicellular communities known as biofilms in response to various stimuli. We find that glycerol, a common carbon source that bacteria can use for energy and biosynthesis, encourages biofilm behaviors such as surface attachment and colony wrinkling by P. aeruginosa. Glycerol can be derived from surfactants that are present in the human lungs, a common infection site. Glycerol-stimulated biofilm phenotypes do not depend on phosphorylation of glycerol but are surprisingly impacted by a glucose breakdown pathway, suggesting that it is glycerol utilization, and not its mere presence or cellular import, that stimulates biofilm phenotypes. Moreover, the same mutations that block glycerol-stimulated biofilm phenotypes also impact P. aeruginosa virulence in both acute and chronic animal models. Notably, a glucose-breakdown mutant (Δedd) counteracts biofilm phenotypes but shows enhanced virulence and stimulates a stronger immune response in Caenorhabditis elegans.
Collapse
Affiliation(s)
- Somalisa Pan
- Department of Microbiology, Oklahoma State University, Stillwater, Oklahoma, USA
| | | | - Christopher W. Hamm
- Department of Microbiology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Mylissa A. Stover
- Department of Biochemistry and Microbiology, OSU Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Daxton R. Butler
- Department of Microbiology, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Crystal A. Shults
- Department of Biochemistry and Microbiology, OSU Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Jacob R. Manjarrez
- Department of Biochemistry and Microbiology, OSU Center for Health Sciences, Tulsa, Oklahoma, USA
| | - Matthew T. Cabeen
- Department of Microbiology, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
50
|
Ciccarelli EJ, Bendelstein M, Yamamoto KK, Reich H, Savage-Dunn C. BMP signaling to pharyngeal muscle in the C. elegans response to a bacterial pathogen regulates anti-microbial peptide expression and pharyngeal pumping. Mol Biol Cell 2024; 35:ar52. [PMID: 38381557 PMCID: PMC11064665 DOI: 10.1091/mbc.e23-05-0185] [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: 05/24/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024] Open
Abstract
Host response to pathogens recruits multiple tissues in part through conserved cell signaling pathways. In Caenorhabditis elegans, the bone morphogenetic protein (BMP) like DBL-1 signaling pathway has a role in the response to infection in addition to other roles in development and postdevelopmental functions. In the regulation of body size, the DBL-1 pathway acts through cell autonomous signal activation in the epidermis (hypodermis). We have now elucidated the tissues that respond to DBL-1 signaling upon exposure to two bacterial pathogens. The receptors and Smad signal transducers for DBL-1 are expressed in pharyngeal muscle, intestine, and epidermis. We demonstrate that expression of receptor-regulated Smad (R-Smad) gene sma-3 in the pharynx is sufficient to improve the impaired survival phenotype of sma-3 mutants and that expression of sma-3 in the intestine has no effect when exposing worms to bacterial infection of the intestine. We also show that two antimicrobial peptide genes - abf-2 and cnc-2 - are regulated by DBL-1 signaling through R-Smad SMA-3 activity in the pharynx. Finally, we show that pharyngeal pumping activity is reduced in sma-3 mutants and that other pharynx-defective mutants also have reduced survival on a bacterial pathogen. Our results identify the pharynx as a tissue that responds to BMP signaling to coordinate a systemic response to bacterial pathogens.
Collapse
Affiliation(s)
- Emma Jo Ciccarelli
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| | | | - Katerina K. Yamamoto
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| | - Hannah Reich
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, CUNY, Flushing, NY 11367
- PhD Program in Biology, The Graduate Center, CUNY, New York, NY 10016
| |
Collapse
|