Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Elife ; 122024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38446031

RESUMO

The survival of hosts during infections relies on their ability to mount effective molecular and behavioral immune responses. Despite extensive research on these defense strategies in various species, including the model organism Caenorhabditis elegans, the neural mechanisms underlying their interaction remain poorly understood. Previous studies have highlighted the role of neural G-protein-coupled receptors (GPCRs) in regulating both immunity and pathogen avoidance, which is particularly dependent on aerotaxis. To address this knowledge gap, we conducted a screen of mutants in neuropeptide receptor family genes. We found that loss-of-function mutations in npr-15 activated immunity while suppressing pathogen avoidance behavior. Through further analysis, NPR-15 was found to regulate immunity by modulating the activity of key transcription factors, namely GATA/ELT-2 and TFEB/HLH-30. Surprisingly, the lack of pathogen avoidance of npr-15 mutant animals was not influenced by oxygen levels. Moreover, our studies revealed that the amphid sensory neuron ASJ is involved in mediating the immune and behavioral responses orchestrated by NPR-15. Additionally, NPR-15 was found to regulate avoidance behavior via the TRPM (transient receptor potential melastatin) gene, GON-2, which may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance. Our study contributes to a broader understanding of host defense strategies and mechanisms underlining the interaction between molecular and behavioral immune responses.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Quimiotaxia , Fatores de Transcrição GATA , Imunidade , Intestinos , Células Receptoras Sensoriais
2.
bioRxiv ; 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-37546751

RESUMO

The survival of hosts during infections relies on their ability to mount effective molecular and behavioral immune responses. Despite extensive research on these defense strategies in various species, including the model organism Caenorhabditis elegans, the neural mechanisms underlying their interaction remain poorly understood. Previous studies have highlighted the role of neural G protein-coupled receptors (GPCRs) in regulating both immunity and pathogen avoidance, which is particularly dependent on aerotaxis. To address this knowledge gap, we conducted a screen of mutants in neuropeptide receptor family genes. We found that loss-of-function mutations in npr-15 activated immunity while suppressing pathogen avoidance behavior. Through further analysis, NPR-15 was found to regulate immunity by modulating the activity of key transcription factors, namely GATA/ELT-2 and TFEB/HLH-30. Surprisingly, the lack of pathogen avoidance of npr-15 mutant animals was not influenced by oxygen levels. Moreover, our studies revealed that the amphid sensory neuron ASJ is involved in mediating the immune and behavioral responses orchestrated by NPR-15. Additionally, NPR-15 was found to regulate avoidance behavior via the TRPM gene, GON-2, which may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance. Our study contributes to a broader understanding of host defense strategies and mechanisms underlining the interaction between molecular and behavioral immune responses.

3.
Front Immunol ; 14: 1093574, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845140

RESUMO

Introduction: The neural control of the immune system by the nervous system is critical to maintaining immune homeostasis, whose disruption may be an underlying cause of several diseases, including cancer, multiple sclerosis, rheumatoid arthritis, and Alzheimer's disease. Methods: Here we studied the role of vagus nerve stimulation (VNS) on gene expression in peripheral blood mononuclear cells (PBMCs). Vagus nerve stimulation is widely used as an alternative treatment for drug-resistant epilepsy. Thus, we studied the impact that VNS treatment has on PBMCs isolated from a cohort of existing patients with medically refractory epilepsy. A comparison of genome-wide changes in gene expression was made between the epilepsy patients treated and non-treated with vagus nerve stimulation. Results: The analysis showed downregulation of genes related to stress, inflammatory response, and immunity, suggesting an anti-inflammatory effect of VNS in epilepsy patients. VNS also resulted in the downregulation of the insulin catabolic process, which may reduce circulating blood glucose. Discussion: These results provide a potential molecular explanation for the beneficial role of the ketogenic diet, which also controls blood glucose, in treating refractory epilepsy. The findings indicate that direct VNS might be a useful therapeutic alternative to treat chronic inflammatory conditions.


Assuntos
Epilepsia Resistente a Medicamentos , Epilepsia , Estimulação do Nervo Vago , Humanos , Criança , Estimulação do Nervo Vago/métodos , Epilepsia Resistente a Medicamentos/terapia , Glicemia , Leucócitos Mononucleares , Epilepsia/terapia , Anti-Inflamatórios
4.
Cell Rep ; 41(5): 111575, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36323254

RESUMO

Alterations in the intestinal physiology caused by pathogen colonization result in immune activation. To provide insights into the mechanisms underlying the control of immune activation by changes in intestinal homeostasis, we conducted a forward genetic screen for suppressors of immune activation by intestinal distension in Caenorhabditis elegans. Our results indicate that C. elegans ACC-4, a member of a family of acetylcholine receptors, is required in immune activation by defects in the defecation motor program or by pathogen infection. ACC-4 acts postsynaptically in non-cholinergic RIM neurons to regulate several immune genes and a Wnt-mediated host immune response. These findings uncover a gut-brain-microbial axis that uses neural cholinergic signaling and the Wnt pathway to control immune activation in response to alterations in intestinal homeostasis.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Receptores Colinérgicos/metabolismo , Via de Sinalização Wnt , Colinérgicos/metabolismo
5.
BMC Biol ; 20(1): 229, 2022 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-36209082

RESUMO

BACKGROUND: Altering animal behavior to reduce pathogen exposure is a key line of defense against pathogen attack. In Caenorhabditis elegans, alterations in intestinal physiology caused by pathogen colonization and sensation of microbial metabolites may lead to activation of pathogen aversive behaviors ranging from aversive reflexes to learned avoidance. However, the neural circuitry between chemosensory neurons that sense pathogenic bacterial cues and the motor neurons responsible for avoidance-associated locomotion remains unknown. RESULTS: Using C. elegans, we found that backward locomotion was a component of learned pathogen avoidance, as animals pre-exposed to Pseudomonas aeruginosa or Enterococcus faecalis showed reflexive aversion to drops of the bacteria driven by chemosensory neurons, including the olfactory AWB neurons. This response also involved intestinal distention and, for E. faecalis, required expression of TRPM channels in the intestine and excretory system. Additionally, we uncovered a circuit composed of olfactory neurons, interneurons, and motor neurons that controls the backward locomotion crucial for learned reflexive aversion to pathogenic bacteria, learned avoidance, and the repulsive odor 2-nonanone. CONCLUSIONS: Using whole-brain simulation and functional assays, we uncovered a novel sensorimotor circuit governing learned reflexive aversion. The discovery of a complete sensorimotor circuit for reflexive aversion demonstrates the utility of using the C. elegans connectome and computational modeling in uncovering new neuronal regulators of behavior.


Assuntos
Proteínas de Caenorhabditis elegans , Canais de Cátion TRPM , Animais , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Pseudomonas aeruginosa , Olfato/fisiologia
6.
Commun Biol ; 5(1): 422, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513700

RESUMO

Innate immune surveillance, which monitors the presence of potentially harmful microorganisms and the perturbations of host physiology that occur in response to infections, is critical to distinguish pathogens from beneficial microbes. Here, we show that multidrug resistance-associated protein-1 (MRP-1) functions in the basolateral membrane of intestinal cells to transport byproducts of cellular redox reactions to control both molecular and behavioral immunity in Caenorhabditis elegans. Pseudomonas aeruginosa infection disrupts glutathione homeostasis, leading to the excess production of the MRP-1 substrate, oxidized glutathione (GSSG). Extracellular GSSG triggers pathogen avoidance behavior and primes naïve C. elegans to induce aversive learning behavior via neural NMDA class glutamate receptor-1 (NMR-1). Our results indicate that MRP-1 transports GSSG, which acts as a danger signal capable of warning C. elegans of changes in intestinal homeostasis, thereby initiating a gut neural signal that elicits an appropriate host defense response.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Aprendizagem da Esquiva , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Dissulfeto de Glutationa , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Oxirredução
7.
PLoS Biol ; 20(3): e3001581, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35263319

RESUMO

Probiotics play a critical role in the control of host intestinal microbial balance, protecting the host from gastrointestinal pathogens, modulating the host immune response, and decreasing host susceptibility to infection. To understand the mechanism underlying the protective effect of probiotics against infections through immune regulation, we examined protection against Salmonella enterica infection following exposure to nonpathogenic Enterococcus faecium in the nematode Caenorhabditis elegans. We found that the transcription factor HLH-26, a REF-1 family member of basic helix-loop-helix transcription factors, was required in the intestine for E. faecium-mediated protection of C. elegans against a lethal S. enterica infection. In addition, we uncovered that defense response genes controlled by the canonical Wnt/BAR-1 pathway were activated upon exposure to E. faecium in an HLH-26-dependent manner. Our findings highlight a role for REF-1/HLH-26 in the control of the Wnt/BAR-1 pathway in probiotic-mediated protection against gut infection.


Assuntos
Proteínas de Caenorhabditis elegans , Probióticos , Infecções por Salmonella , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima
8.
J Biol Chem ; 298(3): 101614, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35101447

RESUMO

Microbial infections have been linked to the onset and severity of neurodegenerative diseases such as amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer's disease, but the underlying mechanisms remain largely unknown. Here, we used a genetic screen for genes involved in protection from infection-associated neurodegeneration and identified the gene mtm-10. We then validated the role of the encoded myotubularin-related protein, MTM-10, in protecting the dendrites of Caenorhabditis elegans from degeneration mediated by oxidative stress or Pseudomonas aeruginosa infection. Further experiments indicated that mtm-10 is expressed in the AWC neurons of C. elegans, where it functions in a cell-autonomous manner to protect the dendrite degeneration caused by pathogen infection. We also confirm that the changes observed in the dendrites of the animals were not because of premature death or overall sickness. Finally, our studies indicated that mtm-10 functions in AWC neurons to preserve chemosensation after pathogen infection. These results reveal an essential role for myotubularin-related protein 10 in the protection of dendrite morphology and function against the deleterious effects of oxidative stress or infection.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Doenças Neurodegenerativas , Neurônios , Proteínas Tirosina Fosfatases não Receptoras , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Dendritos/metabolismo , Dendritos/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo
9.
iScience ; 24(11): 103361, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34765929

RESUMO

[This corrects the article DOI: 10.1016/j.isci.2020.101068.].

10.
Cell Rep ; 35(8): 109187, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34038721

RESUMO

A body of evidence indicates that metazoan immune and aging pathways are largely interconnected, but the mechanisms involved in their homeostatic control remain unclear. In this study, we find that the PITX (paired-like homeodomain) transcription factor UNC-30 controls the tradeoff between immunity and longevity from the nervous system in Caenorhabditis elegans. PITX/UNC-30 functional loss enhances immunity in a GATA/ELT-2- and p38 MAPK/PMK-1-dependent manner and reduced longevity by activating MXD/MDL-1 and the C2H2-type zinc finger transcription factor PQM-1. The immune inhibitory and longevity stimulatory functions of PITX/UNC-30 require the sensory neuron ASG and a signaling pathway controlled by NPR-1, which is a G protein-coupled receptor related to mammalian neuropeptide Y receptors. Our findings uncover a suppressive role of GABAergic signaling in the neural control of a biological tradeoff where energy is allocated toward immunity at the expense of longevity.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunidade Inata/imunologia , Longevidade , Proteínas Nucleares/metabolismo , Pseudomonas aeruginosa/metabolismo , Animais , Fatores de Transcrição/metabolismo
11.
Elife ; 102021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34032213

RESUMO

Upon exposure to harmful microorganisms, hosts engage in protective molecular and behavioral immune responses, both of which are ultimately regulated by the nervous system. Using the nematode Caenorhabditis elegans, we show that ingestion of Enterococcus faecalis leads to a fast pathogen avoidance behavior that results in aversive learning. We have identified multiple sensory mechanisms involved in the regulation of avoidance of E. faecalis. The G-protein coupled receptor NPR-1-dependent oxygen-sensing pathway opposes this avoidance behavior, while an ASE neuron-dependent pathway and an AWB and AWC neuron-dependent pathway are directly required for avoidance. Colonization of the anterior part of the intestine by E. faecalis leads to AWB and AWC mediated olfactory aversive learning. Finally, two transient receptor potential melastatin (TRPM) channels, GON-2 and GTL-2, mediate this newly described rapid pathogen avoidance. These results suggest a mechanism by which TRPM channels may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance and aversive learning by detecting changes in host physiology.


Assuntos
Aprendizagem da Esquiva , Caenorhabditis elegans/microbiologia , Enterococcus faecalis/patogenicidade , Intestinos/microbiologia , Canais de Cátion TRPM/fisiologia , Animais , Enterococcus faecalis/isolamento & purificação , Interações Hospedeiro-Patógeno , Intestinos/patologia , Neurônios/metabolismo , Virulência
12.
PLoS Biol ; 19(3): e3001169, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33788830

RESUMO

The gut-neural axis plays a critical role in the control of several physiological processes, including the communication of signals from the microbiome to the nervous system, which affects learning, memory, and behavior. However, the pathways involved in gut-neural signaling of gut-governed behaviors remain unclear. We found that the intestinal distension caused by the bacterium Pseudomonas aeruginosa induces histone H4 Lys8 acetylation (H4K8ac) in the germline of Caenorhabditis elegans, which is required for both a bacterial aversion behavior and its transmission to the next generation. We show that induction of H4K8ac in the germline is essential for bacterial aversion and that a 14-3-3 chaperone protein family member, PAR-5, is required for H4K8ac. Our findings highlight a role for H4K8ac in the germline not only in the intergenerational transmission of pathogen avoidance but also in the transmission of pathogenic cues that travel through the gut-neural axis to control the aversive behavior.


Assuntos
Microbioma Gastrointestinal/fisiologia , Histonas/genética , Sistema Nervoso/metabolismo , Acetilação , Animais , Aprendizagem da Esquiva/fisiologia , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/metabolismo , Microbioma Gastrointestinal/genética , Células Germinativas/metabolismo , Histonas/metabolismo , Sistema Nervoso/microbiologia , Fenômenos Fisiológicos do Sistema Nervoso/genética , Processamento de Proteína Pós-Traducional , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Transdução de Sinais
13.
Cell Rep ; 31(7): 107662, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32433971

RESUMO

Increasing evidence suggests that deficient immune modulation and microbial infections underline neurodegeneration, but the mechanisms remain obscure. Here, we show that the G-protein-coupled receptor (GPCR) SRBC-48, which belongs to the class BC serpentine receptors, has a protective role in Caenorhabditis elegans dendrite degeneration caused by Pseudomonas aeruginosa infection. Our results indicate that SRBC-48 functions in a cell-autonomous manner in AWC neurons to protect against infection-associated dendrite degeneration. The absence of SRBC-48 results in a reduced lifespan caused by a pathogen infection early in life that induces dendrite degeneration. The decreased longevity in animals deficient in SRBC-48 is due to uncontrolled activation of immune genes, particularly those regulated by the FOXO family transcription factor DAF-16 that is part of the insulin/insulin-like growth factor (IGF)-1 receptor homolog DAF-2. These results reveal how an infection early in life can not only induce dendrite degeneration but also reduce lifespan.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Dendritos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Caenorhabditis elegans , Dendritos/patologia , Longevidade , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa
14.
iScience ; 23(5): 101068, 2020 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-32361270

RESUMO

Cholesterol is an essential nutrient for the function of diverse biological processes and for steroid biosynthesis across metazoans. However, the role of cholesterol in immune function remains understudied. Using the nematode Caenorhabditis elegans, which depends on the external environment for cholesterol, we studied the relationship between cholesterol and innate immunity. We found that the transporter CHUP-1 is required for the effect of cholesterol in the development of innate immunity and that the cholesterol-mediated immune response requires the nuclear hormone receptor NHR-8. Cholesterol acts through NHR-8 to transcriptionally regulate immune genes that are controlled by conserved immune pathways, including a p38/PMK-1 MAPK pathway, a DAF-2/DAF-16 insulin pathway, and an Nrf/SKN-1 pathway. Our results indicate that cholesterol plays a key role in the activation of conserved microbicidal pathways that are essential for survival against bacterial infections.

15.
mBio ; 11(2)2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127446

RESUMO

One of the primary functions of the mucosal barrier, found lining epithelial cells, is to serve as a first-line of defense against microbial pathogens. The major structural components of mucus are heavily glycosylated proteins called mucins. Mucins are key components of the innate immune system as they aid in the clearance of pathogens and can decrease pathogen virulence. It has also been recently reported that individual mucins and derived glycans can attenuate the virulence of the human pathogen Pseudomonas aeruginosa Here, we show data indicating that mucins not only play a role in host defense but that they can also be subverted by P. aeruginosa to cause disease. We found that the mucin MUL-1 and mucin-derived monosaccharides N-acetyl-galactosamine and N-acetylglucosamine are required for P. aeruginosa killing of Caenorhabditis elegans We also found that the defective adhesion of P. aeruginosa to human lung alveolar epithelial cells, deficient in the mucin MUC1, can be reversed by the addition of individual monosaccharides. The monosaccharides identified in this study are found in a wide range of organisms where they act as host factors required for bacterial pathogenesis. While mucins in C. elegans lack sialic acid caps, which makes their monosaccharides readily available, they are capped in other species. Pathogens such as P. aeruginosa that lack sialidases may rely on enzymes from other bacteria to utilize mucin-derived monosaccharides.IMPORTANCE One of the first lines of defense present at mucosal epithelial tissues is mucus, which is a highly viscous material formed by mucin glycoproteins. Mucins serve various functions, but importantly they aid in the clearance of pathogens and debris from epithelial barriers and serve as innate immune factors. In this study, we describe a requirement of host monosaccharides, likely derived from host mucins, for the ability of Pseudomonas aeruginosa to colonize the intestine and ultimately cause death in Caenorhabditis elegans We also demonstrate that monosaccharides alter the ability of bacteria to bind to both Caenorhabditis elegans intestinal cells and human lung alveolar epithelial cells, suggesting that there are conserved mechanisms underlying host-pathogen interactions in a range of organisms. By gaining a better understanding of pathogen-mucin interactions, we can develop better approaches to protect against pathogen infection.


Assuntos
Interações Hospedeiro-Patógeno , Monossacarídeos/metabolismo , Mucinas/metabolismo , Pseudomonas aeruginosa/patogenicidade , Células A549 , Animais , Aderência Bacteriana/efeitos dos fármacos , Caenorhabditis elegans , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Mucina-1/metabolismo , Pseudomonas aeruginosa/fisiologia , Virulência
16.
Curr Opin Neurobiol ; 62: 34-40, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31812835

RESUMO

The nervous and immune systems use bi-directional communication to control host responses against microbial pathogens. Recent studies at the interface of the two systems have highlighted important roles of the nervous system in the regulation of both microbicidal pathways and pathogen avoidance behaviors. Studies on the neural circuits in the simple model host Caenorhabditis elegans have significantly improved our understanding of the roles of conserved neural mechanisms in controlling innate immunity. Moreover, behavioral studies have advanced our understanding of how the nervous system may sense potential pathogens and consequently elicit pathogen avoidance, reducing the risk of infection. In this review, we discuss the neural circuits that regulate both behavioral immunity and molecular immunity in C. elegans.


Assuntos
Caenorhabditis elegans , Animais , Proteínas de Caenorhabditis elegans , Imunidade Inata , Sistema Nervoso , Transdução de Sinais
17.
Bio Protoc ; 10(10): e3623, 2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-33659296

RESUMO

Physical avoidance of pathogens is a crucial defense strategy used by the host to reduce pathogen infection. Hosts display the use of multiple strategies to sense and avoid pathogens, ranging from olfaction to sensing of damage caused by pathogen infection. Understanding various mechanisms of pathogen avoidance has the potential to uncover conserved host defense responses that are important against pathogen infections. Here, we describe protocols for studying pathogen lawn avoidance behavior as well as a change of bacterial preferences in the model nematode Caenorhabditis elegans. Besides, we describe the protocol for measuring preferences for pathogenic and nonpathogenic bacteria after training of the animals on pathogenic bacteria. These assays can be implemented in discovering various mechanisms of host learning that result in the avoidance of pathogens.

18.
Elife ; 82019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31674907

RESUMO

The recognition of pathogens and subsequent activation of defense responses are critical for the survival of organisms. The nematode Caenorhabditis elegans recognizes pathogenic bacteria and elicits defense responses by activating immune pathways and pathogen avoidance. Here we show that chemosensation of phenazines produced by pathogenic Pseudomonas aeruginosa, which leads to rapid activation of DAF-7/TGF-ß in ASJ neurons, is insufficient for the elicitation of pathogen avoidance behavior. Instead, intestinal infection and bloating of the lumen, which depend on the virulence of P. aeruginosa, regulates both pathogen avoidance and aversive learning by modulating not only the DAF-7/TGF-ß pathway but also the G-protein coupled receptor NPR-1 pathway, which also controls aerotaxis behavior. Modulation of these neuroendocrine pathways by intestinal infection serves as a systemic feedback that enables animals to avoid virulent bacteria. These results reveal how feedback from the intestine during infection can modulate the behavior, learning, and microbial perception of the host.


Assuntos
Comportamento Animal , Caenorhabditis elegans/microbiologia , Infecções/metabolismo , Enteropatias/metabolismo , Enteropatias/microbiologia , Aprendizagem , Sistemas Neurossecretores/metabolismo , Transdução de Sinais , Animais , Aprendizagem da Esquiva , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Interações Hospedeiro-Patógeno , Neurônios/metabolismo , Neurônios/microbiologia , Fenazinas , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/fisiologia , Virulência
19.
Curr Opin Immunol ; 60: 30-36, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31117013

RESUMO

Studies in recent years have strengthened the notion that neural mechanisms are involved in the control of immune responses. From initial studies that highlighted the vagus nerve control of inflammatory responses in vertebrates, many advances have been made, including the dissection of specific neural circuits that are involved in controlling immunity. Part of this has been facilitated by the use of a tractable model animal, Caenorhabditis elegans, in which individual neurons involved in sensing pathogens and controlling the immune response have been identified. Importantly, some of the underlying mechanisms involved in the neural control of immune pathways appear to be present in evolutionarily diverse species. This review focuses on some major developments in vertebrates and C. elegans, and how these discoveries may lead to advances in understanding neural-immune connections that govern inflammatory responses.


Assuntos
Neurônios/imunologia , Animais , Humanos , Inflamação/imunologia
20.
mBio ; 10(2)2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31040241

RESUMO

Female Aedes aegypti mosquitoes bite human hosts to obtain a blood meal and, in the process, act as vectors for many disease-causing viruses, including the dengue, chikungunya, yellow fever, and Zika viruses. After a complete meal, the female mosquitoes lose attraction to their hosts for several days. New research shows that pharmacological activation of neuropeptide Y-like receptor (NPYLR) signaling elicits host aversion in female mosquitoes. This behavior of mosquitoes shows remarkable similarities to a bacterial-aversion behavior of the nematode Caenorhabditis elegans Feeding on pathogenic bacteria causes bloating of the gut in C. elegans that leads to activation of NPYLR signaling and bacterial aversion. Several studies suggest that this newly discovered mechanism underlying foraging may be conserved across a large number of species. A better understanding of the regulation of NPYLR signaling pathways could provide molecular targets for the control of eating behaviors in different animals, including human-disease vectors.


Assuntos
Aedes/fisiologia , Comportamento Alimentar , Receptores de Neuropeptídeo Y/metabolismo , Animais , Humanos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...