Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 434
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39000143

RESUMO

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.


Assuntos
Caenorhabditis elegans , Interações Hospedeiro-Patógeno , Imunidade Inata , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Caenorhabditis elegans/microbiologia , Caenorhabditis elegans/imunologia , Pseudomonas aeruginosa/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/imunologia , Humanos , Modelos Animais de Doenças , Virulência
2.
Proc Natl Acad Sci U S A ; 121(29): e2402126121, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38980902

RESUMO

Upon sensing viral RNA, mammalian RIG-I-like receptors (RLRs) activate downstream signals using caspase activation and recruitment domains (CARDs), which ultimately promote transcriptional immune responses that have been well studied. In contrast, the downstream signaling mechanisms for invertebrate RLRs are much less clear. For example, the Caenorhabditis elegans RLR DRH-1 lacks annotated CARDs and up-regulates the distinct output of RNA interference. Here, we found that similar to mammal RLRs, DRH-1 signals through two tandem CARDs (2CARD) to induce a transcriptional immune response. Expression of DRH-1(2CARD) alone in the intestine was sufficient to induce immune gene expression, increase viral resistance, and promote thermotolerance, a phenotype previously associated with immune activation in C. elegans. We also found that DRH-1 is required in the intestine to induce immune gene expression, and we demonstrate subcellular colocalization of DRH-1 puncta with double-stranded RNA inside the cytoplasm of intestinal cells upon viral infection. Altogether, our results reveal mechanistic and spatial insights into antiviral signaling in C. elegans, highlighting unexpected parallels in RLR signaling between C. elegans and mammals.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Transdução de Sinais , Animais , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/imunologia , Transdução de Sinais/imunologia , Intestinos/imunologia , Intestinos/virologia , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/genética , RNA de Cadeia Dupla/metabolismo , RNA de Cadeia Dupla/imunologia , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , RNA Viral/imunologia , RNA Viral/metabolismo , RNA Viral/genética
3.
PLoS Genet ; 20(6): e1011324, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38875298

RESUMO

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


Assuntos
Proteínas Morfogenéticas Ósseas , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Imunidade Inata , Transdução de Sinais , Fator de Crescimento Transformador beta , Animais , Caenorhabditis elegans/microbiologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Imunidade Inata/genética , Ligantes , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Ativinas/metabolismo , Ativinas/genética , Neuropeptídeos
4.
Int J Biol Macromol ; 273(Pt 2): 133206, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38885853

RESUMO

The crude polysaccharide of Bletilla striata in this study was extracted by water extraction and alcohol precipitation and further purified by gel column to yield the purified component Bletilla striata polysaccharide (BSP). Its structure and innate immune regulation activity were studied. BSP mainly comprises mannose and glucose, with a monosaccharide molar ratio of 2.9:1 and a weight-average molecular weight of 28,365 Da. It is a new low-molecular-weight water-soluble neutral glucomannan. BSP contains a â†’ 6)-ß-Manp-(1→, →4)-ß-Glcp-(1→, →4)-ß-Manp-(1 â†’ and →3)-α-Manp-(1 â†’ linear main chain, containing ß-Glcp-(1 â†’ and ß-Manp-(1 â†’ two branched chain fragments were connected to the Man residue at position 4. BSP can enhance the anti-infection ability of Caenorhabditis elegans against Pseudomonas aeruginosa, significantly improve the phagocytic ability of RAW264.7 macrophages, stimulate the secretion of NO and TNF-α, and have good innate immune regulation activity. These findings guide the use of Bletilla striata polysaccharides with immunomodulatory action.


Assuntos
Imunidade Inata , Mananas , Orchidaceae , Animais , Mananas/química , Mananas/farmacologia , Mananas/isolamento & purificação , Camundongos , Orchidaceae/química , Células RAW 264.7 , Imunidade Inata/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/imunologia , Peso Molecular , Pseudomonas aeruginosa/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/química , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Óxido Nítrico/metabolismo , Polissacarídeos/farmacologia , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/química , Agentes de Imunomodulação/isolamento & purificação
5.
STAR Protoc ; 5(2): 103070, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38768031

RESUMO

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.


Assuntos
Caenorhabditis elegans , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Caenorhabditis elegans/microbiologia , Caenorhabditis elegans/imunologia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/imunologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Imunidade Inata
6.
Front Immunol ; 15: 1353747, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38751431

RESUMO

Pathogen avoidance behaviour has been observed across animal taxa as a vital host-microbe interaction mechanism. The nematode Caenorhabditis elegans has evolved multiple diverse mechanisms for pathogen avoidance under natural selection pressure. We summarise the current knowledge of the stimuli that trigger pathogen avoidance, including alterations in aerotaxis, intestinal bloating, and metabolites. We then survey the neural circuits involved in pathogen avoidance, transgenerational epigenetic inheritance of pathogen avoidance, signalling crosstalk between pathogen avoidance and innate immunity, and C. elegans avoidance of non-Pseudomonas bacteria. In this review, we highlight the latest advances in understanding host-microbe interactions and the gut-brain axis.


Assuntos
Caenorhabditis elegans , Interações Hospedeiro-Patógeno , Imunidade Inata , Animais , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Epigênese Genética , Transdução de Sinais , Neurônios/imunologia , Neurônios/metabolismo
7.
J Microbiol Biotechnol ; 34(5): 1109-1118, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38563104

RESUMO

Probiotics, specifically Lacticaseibacillus rhamnosus, have garnered attention for their potential health benefits. This study focuses on evaluating the probiotic properties of candidate probiotics L. rhamnosus IDCC 3201 (3201) using the Caenorhabditis elegans surrogate animal model, a well-established in vivo system for studying host-bacteria interactions. The adhesive ability to the host's gastrointestinal tract is a crucial criterion for selecting potential probiotic bacteria. Our findings demonstrated that 3201 exhibits significantly higher adhesive capabilities compared with Escherichia coli OP50 (OP50), a standard laboratory food source for C. elegans and is comparable with the widely recognized probiotic L. rhamnosus GG (LGG). In lifespan assay, 3201 significantly increased the longevity of C. elegans compared with OP50. In addition, preconditioning with 3201 enhanced C. elegans immune response against four different foodborne pathogenic bacteria. To uncover the molecular basis of these effects, transcriptome analysis elucidated that 3201 modulates specific gene expression related to the innate immune response in C. elegans. C-type lectin-related genes and lysozyme-related genes, crucial components of the immune system, showed significant upregulation after feeding 3201 compared with OP50. These results suggested that preconditioning with 3201 may enhance the immune response against pathogens. Metabolome analysis revealed increased levels of fumaric acid and succinic acid, metabolites of the citric acid cycle, in C. elegans fed with 3201 compared with OP50. Furthermore, there was an increase in the levels of lactic acid, a well-known antimicrobial compound. This rise in lactic acid levels may have contributed to the robust defense mechanisms against pathogens. In conclusion, this study demonstrated the probiotic properties of the candidate probiotic L. rhamnosus IDCC 3201 by using multi-omics analysis.


Assuntos
Caenorhabditis elegans , Lacticaseibacillus rhamnosus , Longevidade , Probióticos , Animais , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Perfilação da Expressão Gênica , Imunidade Inata , Multiômica
8.
Cell Rep ; 43(5): 114138, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678555

RESUMO

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.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Imunidade Inata , Lisossomos , ATPases Vacuolares Próton-Translocadoras , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/imunologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Resposta a Proteínas não Dobradas , ATPases Vacuolares Próton-Translocadoras/metabolismo
9.
G3 (Bethesda) ; 14(5)2024 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-38478633

RESUMO

Innate immunity functions as a rapid defense against broad classes of pathogenic agents. While the mechanisms of innate immunity in response to antigen exposure are well-studied, how pathogen exposure activates the innate immune responses and the role of genetic variation in immune activity is currently being investigated. Previously, we showed significant survival differences between the N2 and the CB4856 Caenorhabditis elegans isolates in response to Staphylococcus epidermidis infection. One of those differences was expression of the mab-5 Hox family transcription factor, which was induced in N2, but not CB4856, after infection. In this study, we use survival assays and RNA-sequencing to better understand the role of mab-5 in response to S. epidermidis. We found that mab-5 loss-of-function (LOF) mutants were more susceptible to S. epidermidis infection than N2 or mab-5 gain-of-function (GOF) mutants, but not as susceptible as CB4856 animals. We then conducted transcriptome analysis of infected worms and found considerable differences in gene expression profiles when comparing animals with mab-5 LOF to either N2 or mab-5 GOF. N2 and mab-5 GOF animals showed a significant enrichment in expression of immune genes and C-type lectins, whereas mab-5 LOF mutants did not. Overall, gene expression profiling in mab-5 mutants provided insight into MAB-5 regulation of the transcriptomic response of C. elegans to pathogenic bacteria and helps us to understand mechanisms of innate immune activation and the role that transcriptional regulation plays in organismal health.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Proteínas de Homeodomínio , Imunidade Inata , Staphylococcus epidermidis , Fatores de Transcrição , Animais , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/imunologia , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mutação , Infecções Estafilocócicas/imunologia , Staphylococcus epidermidis/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
10.
Environ Pollut ; 314: 120294, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36181932

RESUMO

Per- and Poly-fluoroalkyl substances (PFAS) are major persistent environmental contaminants. Epidemiological studies have linked PFAS exposures to altered immunity and increased occurrence of infections in children. However, the mechanisms leading to immune susceptibility to bacterial infections remains unclear. To elucidate the mechanism, transcriptional alteration in the Caenorhabditis elegans model caused by a PFAS contaminated environmental water and two reconstituted PFAS solutions were evaluated using RNA-sequencing. PFAS affected the expression of several genes involved in C. elegans immune surveillance to Gram-positive bacteria (cpr-2, tag-38, spp-1, spp-5, clec-7, clec-172). The combined exposure to PFAS and Staphylococcus aureus significantly reduced C. elegans survival and increased intestinal membrane permeability. Furthermore, the growth of S. aureus in the presence of PFAS increased the expression of virulence genes, specifically, the virulence gene regulator saeR and α-hemolysin, hla, which resulted in increased hemolytic activity. The present study demonstrated that PFAS exposure not only increased C. elegans susceptibility to pathogens by reducing host immunity and increasing intestinal membrane permeability, but also increased bacteria virulence. This presents a broader implication for humans and other animals, where environmental contaminants simultaneously reduce host resilience, while, increasing microbial pathogenicity.


Assuntos
Caenorhabditis elegans , Fluorocarbonos , Staphylococcus aureus , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Fluorocarbonos/toxicidade , Proteínas Hemolisinas , Imunidade , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade , Virulência/genética , Poluentes Ambientais/toxicidade
11.
New Microbiol ; 45(1): 51-61, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35403847

RESUMO

Candida albicans can cause infections ranging from superficial skin infections to life-threateningsystemic infections in immunocompromised hosts. Although several C. albicans virulence factorsare widely discussed in great detail, intrinsic host determinants that are critical for C. albicanspathogenesis remain less interested and poorly understood. In view of this, a model of Caenorhabditiselegans was used to study host longevity and immunity in response to C. albicans pathogenesis.The influence of C. albicans in pathological and survival aspects was evaluated using C. elegans.C. albicans hyphal formation in different C. elegans genetic backgrounds was evaluated. Moreover,several C. elegans fluorescent proteins as gene expression markers upon C. albicans infectionswere evaluated. C. albicans is pathogenic to C. elegans and reduces the lifespan of C. elegans inassociation with repression of the insulin/IGF-1-like signaling (IIS) pathway. Moreover, repressionof DAF-16/forkhead transcription factor increases aggressiveness of C. albicans by enhancing hyphalformation. In addition, infection of C. albicans increases lipofuscin accumulation, promotes DAF-16nuclear translocation, increases superoxide dismutase (SOD-3) expression, which coordinately linksbetween aging and innate immunity. Thus, we demonstrate here the strategy to utilize C. elegans asa model host to elucidate host genetic determinants that provide insights into the pathogenesis ofC. albicans infections.


Assuntos
Proteínas de Caenorhabditis elegans , Candidíase , Fatores de Transcrição Forkhead , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Candida albicans , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Imunidade Inata , Insulina/genética , Insulina/metabolismo , Longevidade/genética , Mutação
12.
Nat Commun ; 13(1): 17, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013162

RESUMO

Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Caenorhabditis elegans , Enterócitos , Interações Hospedeiro-Patógeno/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Caenorhabditis elegans/virologia , Proteínas de Caenorhabditis elegans/imunologia , Proteínas de Caenorhabditis elegans/metabolismo , Enterócitos/imunologia , Enterócitos/microbiologia , Enterócitos/virologia , Imunidade Inata/fisiologia , Intestinos/microbiologia , Intestinos/virologia , Invertebrados/imunologia , Microsporídios/patogenicidade , Vírus de RNA/patogenicidade
13.
Front Immunol ; 12: 744454, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804026

RESUMO

Innate immunity is the first line of host defense against pathogen infection in metazoans. However, the molecular mechanisms of the complex immune regulatory network are not fully understood. Based on a transcriptome profiling of the nematode Caenorhabditis elegans, we found that a bZIP transcription factor ZIP-11 was up-regulated upon Pseudomonas aeruginosa PA14 infection. The tissue specific RNAi knock-down and rescue data revealed that ZIP-11 acts in intestine to promote host resistance against P. aeruginosa PA14 infection. We further showed that intestinal ZIP-11 regulates innate immune response through constituting a feedback loop with the conserved PMK-1/p38 mitogen-activated protein signaling pathway. Intriguingly, ZIP-11 interacts with a CCAAT/enhancer-binding protein, CEBP-2, to mediate the transcriptional response to P. aeruginosa PA14 infection independently of PMK-1/p38 pathway. In addition, human homolog ATF4 can functionally substitute for ZIP-11 in innate immune regulation of C. elegans. Our findings indicate that the ZIP-11/ATF4 genetic program activates local innate immune response through conserved PMK-1/p38 and CEBP-2/C/EBPγ immune signals in C. elegans, raising the possibility that a similar process may occur in other organisms.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Proteínas de Caenorhabditis elegans/imunologia , Imunidade Inata/imunologia , Fator 4 Ativador da Transcrição/imunologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/imunologia , Humanos
14.
mBio ; 12(5): e0257921, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34634942

RESUMO

A variety of effector proteins contribute to host defense in Caenorhabditis elegans. However, beyond lytic enzymes and antimicrobial peptides and proteins, little is known about the exact function of these infection-related effectors. This study set out to identify pathogen-dependent cytokine-like molecules, focusing on C-type lectin domain-containing proteins (CLECs). In total, 38 CLECs that are differentially regulated in response to bacterial infections have been previously identified by microarray and transcriptome sequencing (RNA-seq) analyses in C. elegans. We successfully cloned 18 of these 38 CLECs and chose to focus on CLEC-47 because, among these 18 cloned CLECs, it was the smallest protein and was recombinantly expressed at the highest levels in prokaryotic cells examined by SDS-PAGE. Quantitative real-time PCR (qRT-PCR/qPCR) showed that the expression of clec-47 was induced by a variety of Gram-positive bacterial pathogens, including Enterococcus faecium, Staphylococcus aureus, and Cutibacterium acnes, but was suppressed by the Gram-negative bacteria Klebsiella pneumoniae and Pseudomonas aeruginosa. By expressing CLEC-47 in HEK 293 cells, we showed that CLEC-47 is released into the culture media, which the Golgi apparatus inhibitors (brefeldin A [BFA] and GolgiStop) could block. Purified recombinant CLEC-47 (maltose binding protein [MBP]-CLEC-47-His) did not display antimicrobial activity against ESKAPE pathogen isolates but bound directly to murine macrophage J774A.1 cells. Recombinant CLEC-47 attracted and recruited J774A.1 cells in a chemotaxis assay. In addition, qPCR studies and enzyme-linked immunosorbent assays (ELISAs) showed that CLEC-47 activates J774A.1 cells in a dose- and time-dependent manner to express the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6, and Macrophage Inflammatory Protein 2 (MIP-2). Moreover, C. elegans, fed with CLEC-47-expressing Escherichia coli, demonstrated enhanced expression of several antimicrobial proteins (CNC-1, CNC-2, CPR-1, and CPR-2) as well as the detoxification protein MTL-1. These data suggest that CLEC-47 functions as a novel cytokine-like signaling molecule and exemplify how the study of infection-related effectors in C. elegans can help elucidate the evolution of immune responses. IMPORTANCE A variety of effector proteins contribute to host defense in the nematode Caenorhabditis elegans. However, little is known about the exact function of these infection-related effectors beyond lytic enzymes and antimicrobial peptides and proteins. This study set out to identify pathogen-dependent cytokine-like molecules, and we focus on the C-type lectin domain-containing proteins (CLECs). Our data suggest that CLEC-47 functions as a novel cytokine-like signaling molecule and exemplify how the study of infection-related effectors in nematodes can help elucidate the evolution of immune responses.


Assuntos
Infecções Bacterianas/imunologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/imunologia , Caenorhabditis elegans/química , Caenorhabditis elegans/imunologia , Citocinas/imunologia , Imunidade Inata , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiologia , Linhagem Celular , Citocinas/classificação , Citocinas/genética , Células HEK293 , Humanos , Camundongos , Domínios Proteicos
15.
Int J Mol Sci ; 22(19)2021 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-34639082

RESUMO

Pseudomonas donghuensis HYS is more virulent than P. aeruginosa toward Caenorhabditis elegans but the mechanism underlying virulence is unclear. This study is the first to report that the specific gene cluster gtrA/B/II in P. donghuensis HYS is involved in the virulence of this strain toward C. elegans, and there are no reports of GtrA, GtrB and GtrII in any Pseudomonas species. The pathogenicity of P. donghuensis HYS was evaluated using C. elegans as a host. Based on the prediction of virulence factors and comparative genomic analysis of P. donghuensis HYS, we identified 42 specific virulence genes in P. donghuensis HYS. Slow-killing assays of these genes showed that the gtrAB mutation had the greatest effect on the virulence of P. donghuensis HYS, and GtrA, GtrB and GtrII all positively affected P. donghuensis HYS virulence. Two critical GtrII residues (Glu47 and Lys480) were identified in P. donghuensis HYS. Transmission electron microscopy (TEM) showed that GtrA, GtrB and GtrII were involved in the glucosylation of lipopolysaccharide (LPS) O-antigen in P. donghuensis HYS. Furthermore, colony-forming unit (CFU) assays showed that GtrA, GtrB and GtrII significantly enhanced P. donghuensis HYS colonization in the gut of C. elegans, and glucosylation of LPS O-antigen and colonization in the host intestine contributed to the pathogenicity of P. donghuensis HYS. In addition, experiments using the worm mutants ZD101, KU4 and KU25 revealed a correlation between P. donghuensis HYS virulence and the TIR-1/SEK-1/PMK-1 pathways of the innate immune p38 MAPK pathway in C. elegans. In conclusion, these results reveal that the specific virulence gene cluster gtrA/B/II contributes to the unique pathogenicity of HYS compared with other pathogenic Pseudomonas, and that this process also involves C. elegans innate immunity. These findings significantly increase the available information about GtrA/GtrB/GtrII-based virulence mechanisms in the genus Pseudomonas.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/imunologia , Imunidade Inata/imunologia , Família Multigênica , Pseudomonas/patogenicidade , Fatores de Virulência/metabolismo , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Virulência , Fatores de Virulência/genética
16.
Genesis ; 59(12): e23454, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34664387

RESUMO

In the recent past, Caenorhabditis elegans has emerged as one of the leading nematode models for studying host-microbe interactions on molecular, cellular, or organismal levels. In general, morphological and functional similarities of the gut of C. elegans with respect to that of human has brought in speculations on the study of the intestinal microbiota. On the other hand, probiotics have proved their efficacy in metabolism, development, and pathogenesis thereby inducing an immune response in C. elegans. Nurturing C. elegans with probiotics has led to immunomodulatory effects in the intestinal microbiota, proposing C. elegans as one of the in vivo screening criteria to select potential probiotic bacteria for host health-promoting factors. The major prospect of these probiotics is to exert longevity toward the host in diverse environmental conditions. The extent of research on probiotic metabolism has shed light on mechanisms of the immunomodulatory effect exerted by the nematode model. This review discusses various aspects of the effects of probiotics in improving the health and mechanisms involved in conferring immunity in C. elegans.


Assuntos
Caenorhabditis elegans/microbiologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata/imunologia , Probióticos , Animais , Caenorhabditis elegans/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Longevidade/imunologia
17.
Biochem Soc Trans ; 49(5): 2307-2317, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34623403

RESUMO

The simple notion 'infection causes an immune response' is being progressively refined as it becomes clear that immune mechanisms cannot be understood in isolation, but need to be considered in a more global context with other cellular and physiological processes. In part, this reflects the deployment by pathogens of virulence factors that target diverse cellular processes, such as translation or mitochondrial respiration, often with great molecular specificity. It also reflects molecular cross-talk between a broad range of host signalling pathways. Studies with the model animal C. elegans have uncovered a range of examples wherein innate immune responses are intimately connected with different homeostatic mechanisms, and can influence reproduction, ageing and neurodegeneration, as well as various other aspects of its biology. Here we provide a short overview of a number of such connections, highlighting recent discoveries that further the construction of a fully integrated view of innate immunity.


Assuntos
Caenorhabditis elegans/imunologia , Imunidade Inata , Animais , Caenorhabditis elegans/metabolismo , Oomicetos/fisiologia , Transdução de Sinais , Sono
18.
Life Sci Alliance ; 4(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34583931

RESUMO

The mitochondrial unfolded protein response (mitoUPR) is an evolutionarily conserved pathway that responds to mitochondria insults through transcriptional changes, mediated by the transcription factor ATFS-1/ATF-5, which acts to restore mitochondrial homeostasis. In this work, we characterized the role of ATFS-1 in responding to organismal stress. We found that activation of ATFS-1 is sufficient to cause up-regulation of genes involved in multiple stress response pathways including the DAF-16-mediated stress response pathway, the cytosolic unfolded protein response, the endoplasmic reticulum unfolded protein response, the SKN-1-mediated oxidative stress response pathway, the HIF-1-mediated hypoxia response pathway, the p38-mediated innate immune response pathway, and antioxidant genes. Constitutive activation of ATFS-1 increases resistance to multiple acute exogenous stressors, whereas disruption of atfs-1 decreases stress resistance. Although ATFS-1-dependent genes are up-regulated in multiple long-lived mutants, constitutive activation of ATFS-1 decreases lifespan in wild-type animals. Overall, our work demonstrates that ATFS-1 serves a vital role in organismal survival of acute stressors through its ability to activate multiple stress response pathways but that chronic ATFS-1 activation is detrimental for longevity.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Mitocôndrias/metabolismo , Transdução de Sinais/genética , Estresse Fisiológico/genética , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas/genética , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/imunologia , Proteínas de Caenorhabditis elegans/genética , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Imunidade Inata , Longevidade/genética , Mutação , Estresse Oxidativo/genética , Transdução de Sinais/imunologia , Estresse Fisiológico/imunologia , Fatores de Transcrição/genética , Regulação para Cima/genética
19.
Cell Rep ; 36(7): 109529, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34407394

RESUMO

In Caenorhabditis elegans, ROS generated in response to intestinal infection induces SKN-1, a protective transcription factor homologous to nuclear factor erythroid 2-related factor 1 or 2 (NRF1/2) in mammals. Many factors regulate SKN-1, including the p38 mitogen-activated protein kinase (MAPK) cascade that activates SKN-1 by phosphorylation. In this work, another positive regulator of SKN-1 is identified: NIPI-3, a Tribbles pseudokinase. NIPI-3 has been reported to protect against intestinal infection by negatively regulating the CCAT enhancer binding protein (C/EBP) bZIP transcription factor CEBP-1. Here we demonstrate that CEBP-1 positively regulates the vhp-1 transcript, which encodes a phosphatase that dephosphorylates the p38 MAPK called PMK-1. The increased levels of VHP-1 caused by CEBP-1 transcriptional enhancement result in less PMK-1 phosphorylation, affecting SKN-1 activity and intestinal resistance to the pathogen. The data support a model in which NIPI-3's negative regulation of CEBP-1 decreases VHP-1 phosphatase activity, allowing increased stimulation of SKN-1 activity by the p38 MAPK phosphorylation cascade in the intestine.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/imunologia , Proteínas de Ligação a DNA/metabolismo , Imunidade Inata , Intestinos/imunologia , Proteínas Quinases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Proteínas Quinases/genética
20.
Genes (Basel) ; 12(8)2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34440335

RESUMO

The nematode Caenorhabditis elegans has been a versatile model for understanding the molecular responses to abiotic stress and pathogens. In particular, the response to heat stress and virus infection has been studied in detail. The Orsay virus (OrV) is a natural virus of C. elegans and infection leads to intracellular infection and proteostatic stress, which activates the intracellular pathogen response (IPR). IPR related gene expression is regulated by the genes pals-22 and pals-25, which also control thermotolerance and immunity against other natural pathogens. So far, we have a limited understanding of the molecular responses upon the combined exposure to heat stress and virus infection. We test the hypothesis that the response of C. elegans to OrV infection and heat stress are co-regulated and may affect each other. We conducted a combined heat-stress-virus infection assay and found that after applying heat stress, the susceptibility of C. elegans to OrV was decreased. This difference was found across different wild types of C. elegans. Transcriptome analysis revealed a list of potential candidate genes associated with heat stress and OrV infection. Subsequent mutant screens suggest that pals-22 provides a link between viral response and heat stress, leading to enhanced OrV tolerance of C. elegans after heat stress.


Assuntos
Caenorhabditis elegans/virologia , Resposta ao Choque Térmico , Nodaviridae/patogenicidade , Infecções por Vírus de RNA/imunologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Suscetibilidade a Doenças , Genes de Helmintos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...