Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38712048

RESUMEN

Background & Aims: Despite increasing therapeutic options in the treatment of ulcerative colitis (UC), achieving disease remission remains a major clinical challenge. Nonresponse to therapy is common and clinicians have little guidance in selecting the optimal therapy for an individual patient. This study examined whether patient-derived materials could predict individual clinical responsiveness to the Janus kinase (JAK) inhibitor, tofacitinib, prior to treatment initiation. Method: In 48 patients with UC initiating tofacitinib, we longitudinally collected clinical covariates, stool, and colonic biopsies to analyze the microbiota, transcriptome, and exome variations associated with clinical responsiveness at week 24. We established patient-derived organoids (n = 23) to determine how their viability upon stimulation with proinflammatory cytokines in the presence of tofacitinib related to drug responsiveness in patients. We performed additional biochemical analyses of organoids and primary tissues to identify the mechanism underlying differential tofacitinib sensitivity. Results: The composition of the gut microbiota, rectal transcriptome, inflammatory biomarkers, and exome variations were indistinguishable among UC patients prior to tofacitinib treatment. However, a subset of patient-derived organoids displayed reduced sensitivity to tofacitinib as determined by the ability of the drug to inhibit STAT1 phosphorylation and loss of viability upon cytokine stimulation. Remarkably, sensitivity of organoids to tofacitinib predicted individual clinical patient responsiveness. Reduced responsiveness to tofacitinib was associated with decreased levels of the cationic transporter MATE1, which mediates tofacitinib uptake. Conclusions: Patient-derived intestinal organoids predict and identify mechanisms of individual tofacitinib responsiveness in UC. Specifically, MATE1 expression predicted clinical response to tofacitinib.

2.
Cell Host Microbe ; 31(9): 1450-1468.e8, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37652008

RESUMEN

Loss of antimicrobial proteins such as REG3 family members compromises the integrity of the intestinal barrier. Here, we demonstrate that overproduction of REG3 proteins can also be detrimental by reducing a protective species in the microbiota. Patients with inflammatory bowel disease (IBD) experiencing flares displayed heightened levels of secreted REG3 proteins that mediated depletion of Enterococcus faecium (Efm) from the gut microbiota. Efm inoculation of mice ameliorated intestinal inflammation through activation of the innate immune receptor NOD2, which was associated with the bacterial DL-endopeptidase SagA that generates NOD2-stimulating muropeptides. NOD2 activation in myeloid cells induced interleukin-1ß (IL-1ß) secretion to increase the proportion of IL-22-producing CD4+ T helper cells and innate lymphoid cells that promote tissue repair. Finally, Efm was unable to protect mice carrying a NOD2 gene variant commonly found in IBD patients. Our findings demonstrate that inflammation self-perpetuates by causing aberrant antimicrobial activity that disrupts symbiotic relationships with gut microbes.


Asunto(s)
Antiinfecciosos , Enterococcus faecium , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Inmunidad Innata , Linfocitos , Inflamación
3.
bioRxiv ; 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36778381

RESUMEN

Loss of antimicrobial proteins such as REG3 family members compromises the integrity of the intestinal barrier. Here, we demonstrate that overproduction of REG3 proteins can also be detrimental by reducing a protective species in the microbiota. Patients with inflammatory bowel disease (IBD) experiencing flares displayed heightened levels of secreted REG3 proteins that mediated depletion of Enterococcus faecium ( Efm ) from the gut microbiota. Efm inoculation of mice ameliorated intestinal inflammation through activation of the innate immune receptor NOD2, which was associated with the bacterial DL-endopeptidase SagA. Microbiota sensing by NOD2 in myeloid cells mediated IL-1ß secretion and increased the proportion of IL-22-producing CD4 + T helper cells and innate lymphoid cells. Finally, Efm was unable to protect mice carrying a NOD2 gene variant commonly found in IBD patients. Our findings demonstrate that inflammation self-perpetuates by causing aberrant antimicrobial activity that disrupts symbiotic relationships with gut microbes.

4.
PLoS Biol ; 20(3): e3001592, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35358182

RESUMEN

Gastrointestinal effects associated with Coronavirus Disease 2019 (COVID-19) are highly variable for reasons that are not understood. In this study, we used intestinal organoid-derived cultures differentiated from primary human specimens as a model to examine interindividual variability. Infection of intestinal organoids derived from different donors with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) resulted in orders of magnitude differences in virus replication in small intestinal and colonic organoid-derived monolayers. Susceptibility to infection correlated with angiotensin I converting enzyme 2 (ACE2) expression level and was independent of donor demographic or clinical features. ACE2 transcript levels in cell culture matched the amount of ACE2 in primary tissue, indicating that this feature of the intestinal epithelium is retained in the organoids. Longitudinal transcriptomics of organoid-derived monolayers identified a delayed yet robust interferon signature, the magnitude of which corresponded to the degree of SARS-CoV-2 infection. Interestingly, virus with the Omicron variant spike (S) protein infected the organoids with the highest infectivity, suggesting increased tropism of the virus for intestinal tissue. These results suggest that heterogeneity in SARS-CoV-2 replication in intestinal tissues results from differences in ACE2 levels, which may underlie variable patient outcomes.


Asunto(s)
COVID-19 , Enzima Convertidora de Angiotensina 2/genética , Humanos , Organoides , SARS-CoV-2
5.
Immunity ; 55(2): 237-253.e8, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35081371

RESUMEN

The Th17 cell-lineage-defining cytokine IL-17A contributes to host defense and inflammatory disease by coordinating multicellular immune responses. The IL-17 receptor (IL-17RA) is expressed by diverse intestinal cell types, and therapies targeting IL-17A induce adverse intestinal events, suggesting additional tissue-specific functions. Here, we used multiple conditional deletion models to identify a role for IL-17A in secretory epithelial cell differentiation in the gut. Paneth, tuft, goblet, and enteroendocrine cell numbers were dependent on IL-17A-mediated induction of the transcription factor ATOH1 in Lgr5+ intestinal epithelial stem cells. Although dispensable at steady state, IL-17RA signaling in ATOH1+ cells was required to regenerate secretory cells following injury. Finally, IL-17A stimulation of human-derived intestinal organoids that were locked into a cystic immature state induced ATOH1 expression and rescued secretory cell differentiation. Our data suggest that the cross talk between immune cells and stem cells regulates secretory cell lineage commitment and the integrity of the mucosa.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Mucosa Intestinal/citología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina-17/metabolismo , Células Madre/metabolismo , Animales , Comunicación Celular , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran/efectos adversos , Humanos , Interleucina-17/metabolismo , Interleucina-17/farmacología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Intestinos/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Receptores de Interleucina-17/deficiencia , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Células Madre/citología
6.
Viruses ; 13(12)2021 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-34960706

RESUMEN

Epidemic RNA viruses seem to arise year after year leading to countless infections and devastating disease. SARS-CoV-2 is the most recent of these viruses, but there will undoubtedly be more to come. While effective SARS-CoV-2 vaccines are being deployed, one approach that is still missing is effective antivirals that can be used at the onset of infections and therefore prevent pandemics. Here, we screened FDA-approved compounds against SARS-CoV-2. We found that atovaquone, a pyrimidine biosynthesis inhibitor, is able to reduce SARS-CoV-2 infection in human lung cells. In addition, we found that berberine chloride, a plant-based compound used in holistic medicine, was able to inhibit SARS-CoV-2 infection in cells through direct interaction with the virion. Taken together, these studies highlight potential avenues of antiviral development to block emerging viruses. Such proactive approaches, conducted well before the next pandemic, will be essential to have drugs ready for when the next emerging virus hits.


Asunto(s)
Antivirales/farmacología , Atovacuona/farmacología , Berberina/farmacología , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Células Epiteliales Alveolares , Animales , Berberina/química , Proliferación Celular/efectos de los fármacos , Cloruros/química , Cloruros/farmacología , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Proguanil/farmacología , Células Vero , Virión/efectos de los fármacos
7.
Cell Host Microbe ; 28(6): 789-797.e5, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33301718

RESUMEN

The colonic microbiota exhibits cross-sectional heterogeneity, but the mechanisms that govern its spatial organization remain incompletely understood. Here we used Citrobacter rodentium, a pathogen that colonizes the colonic surface, to identify microbial traits that license growth and survival in this spatial niche. Previous work showed that during colonic crypt hyperplasia, type III secretion system (T3SS)-mediated intimate epithelial attachment provides C. rodentium with oxygen for aerobic respiration. However, we find that prior to the development of colonic crypt hyperplasia, T3SS-mediated intimate attachment is not required for aerobic respiration but for hydrogen peroxide (H2O2) respiration using cytochrome c peroxidase (Ccp). The epithelial NADPH oxidase NOX1 is the primary source of luminal H2O2 early after C. rodentium infection and is required for Ccp-dependent growth. Our results suggest that NOX1-derived H2O2 is a resource that governs bacterial growth and survival in close proximity to the mucosal surface during gut homeostasis.


Asunto(s)
Citrobacter rodentium/crecimiento & desarrollo , Citrobacter rodentium/metabolismo , Citocromo-c Peroxidasa/fisiología , Peróxido de Hidrógeno/metabolismo , NADPH Oxidasa 1/fisiología , Anaerobiosis , Animales , Colon/microbiología , ADN Bacteriano , Heces/microbiología , Femenino , Vida Libre de Gérmenes , Homeostasis , Interacciones Huésped-Patógeno , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Ribosómico 16S , Organismos Libres de Patógenos Específicos , Sistemas de Secreción Tipo III/fisiología
8.
mBio ; 11(4)2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32723914

RESUMEN

A multifunctional autoprocessing repeats-in-toxin (MARTX) toxin plays an essential role in the virulence of many pathogens, including a fulminating human pathogen Vibrio vulnificus H-NS and HlyU repress and derepress expression of the MARTX toxin gene rtxA in V. vulnificus, respectively. However, little is known about other regulatory proteins and environmental signals involved in rtxA regulation. In this study, we found that a leucine-responsive regulatory protein (Lrp) activates rtxA by binding directly and specifically to the rtxA promoter, P rtxA Phased hypersensitivity resulting from DNase I cleavage of the P rtxA regulatory region suggests that Lrp probably induces DNA bending in P rtxA Lrp activates P rtxA independently of H-NS and HlyU, and leucine inhibits Lrp binding to P rtxA and reduces the Lrp-mediated activation. Furthermore, a cyclic AMP receptor protein (CRP) represses P rtxA , and exogenous glucose relieves the CRP-mediated repression. Biochemical and mutational analyses demonstrated that CRP binds directly and specifically to the upstream region of P rtxA , which presumably alters the DNA conformation in P rtxA and thus represses rtxA Moreover, CRP represses expression of lrp and hlyU by binding directly to their upstream regions, forming coherent feed-forward loops with Lrp and HlyU. In conclusion, expression of rtxA is controlled by a regulatory network comprising CRP, Lrp, H-NS, and HlyU in response to changes in host environmental signals such as leucine and glucose. This collaborative regulation enables the elaborate expression of rtxA, thereby enhancing the fitness and pathogenesis of V. vulnificus during the course of infection.IMPORTANCE A MARTX toxin, RtxA, is an essential virulence factor of many pathogens, including Vibrio species. H-NS and HlyU repress and derepress, respectively, rtxA expression of a life-threatening pathogen, Vibrio vulnificus We found that Lrp directly activates rtxA independently of H-NS and HlyU, and leucine inhibits the Lrp-mediated activation of rtxA Furthermore, we demonstrated that CRP represses rtxA but derepresses in the presence of exogenous glucose. CRP represses rtxA not only directly by binding to upstream of rtxA but also indirectly by repressing lrp and hlyU This is the first report of a regulatory network comprising CRP, Lrp, H-NS, and HlyU, which coordinates the rtxA expression in response to environmental signals such as leucine and glucose during infection. This elaborate regulatory network will enhance the fitness of V. vulnificus and contribute to its successful infection within the host.


Asunto(s)
Toxinas Bacterianas/genética , Proteína Receptora de AMP Cíclico/genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Vibrio vulnificus/genética , Proteína Receptora de AMP Cíclico/metabolismo , Ambiente , Glucosa/farmacología , Humanos , Proteína Reguladora de Respuesta a la Leucina/genética , Proteína Reguladora de Respuesta a la Leucina/metabolismo , Regiones Promotoras Genéticas , Vibriosis/microbiología , Vibrio vulnificus/efectos de los fármacos , Vibrio vulnificus/patogenicidad , Virulencia , Factores de Virulencia
9.
J Biol Chem ; 295(16): 5350-5361, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32169898

RESUMEN

For successful infection of their hosts, pathogenic bacteria recognize host-derived signals that induce the expression of virulence factors in a spatiotemporal manner. The fulminating food-borne pathogen Vibrio vulnificus produces a cytolysin/hemolysin protein encoded by the vvhBA operon, which is a virulence factor preferentially expressed upon exposure to murine blood and macrophages. The Fe-S cluster containing transcriptional regulator IscR activates the vvhBA operon in response to nitrosative stress and iron starvation, during which the cellular IscR protein level increases. Here, electrophoretic mobility shift and DNase I protection assays revealed that IscR directly binds downstream of the vvhBA promoter P vvhBA , which is unusual for a positive regulator. We found that in addition to IscR, the transcriptional regulator HlyU activates vvhBA transcription by directly binding upstream of P vvhBA , whereas the histone-like nucleoid-structuring protein (H-NS) represses vvhBA by extensively binding to both downstream and upstream regions of its promoter. Of note, the binding sites of IscR and HlyU overlapped with those of H-NS. We further substantiated that IscR and HlyU outcompete H-NS for binding to the P vvhBA regulatory region, resulting in the release of H-NS repression and vvhBA induction. We conclude that concurrent antirepression by IscR and HlyU at regions both downstream and upstream of P vvhBA provides V. vulnificus with the means of integrating host-derived signal(s) such as nitrosative stress and iron starvation for precise regulation of vvhBA transcription, thereby enabling successful host infection.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Deficiencias de Hierro , Nitrógeno/metabolismo , Operón , Estrés Fisiológico , Vibrio vulnificus/genética , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Células Cultivadas , Hierro/metabolismo , Ratones , Regiones Promotoras Genéticas , Células RAW 264.7 , Factores de Transcripción/metabolismo , Vibrio vulnificus/metabolismo
10.
Sci Rep ; 9(1): 4346, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30867441

RESUMEN

Increasing antibiotic resistance has led to the development of new strategies to combat bacterial infection. Anti-virulence strategies that impair virulence of bacterial pathogens are one of the novel approaches with less selective pressure for developing resistance than traditional strategies that impede viability. In this study, a small molecule CM14 [N-(4-oxo-4H-thieno[3,4-c]chromen-3-yl)-3-phenylprop-2-ynamide] that inhibits the activity of HlyU, a transcriptional regulator essential for the virulence of the fulminating human pathogen Vibrio vulnificus, has been identified. Without affecting bacterial growth or triggering the host cell death, CM14 reduces HlyU-dependent expression of virulence genes in V. vulnificus. In addition to the decreased hemolysis of human erythrocytes, CM14 impedes host cell rounding and lysis caused by V. vulnificus. Notably, CM14 significantly enhances survival of mice infected with V. vulnificus by alleviating hepatic and renal dysfunction and systemic inflammation. Biochemical, mass spectrometric, and mutational analyses revealed that CM14 inhibits HlyU from binding to target DNA by covalently modifying Cys30. Remarkably, CM14 decreases the expression of various virulence genes of other Vibrio species and thus attenuates their virulence phenotypes. Together, this molecule could be an anti-virulence agent against HlyU-harboring Vibrio species with a low selective pressure for the emergence of resistance.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Vibrio vulnificus/patogenicidad , Virulencia/efectos de los fármacos , Animales , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Ratones , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo , Factores de Virulencia/genética
11.
Front Immunol ; 9: 2095, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283443

RESUMEN

T helper type 17 (Th17) cells are a subset of pro-inflammatory T helper cells that mediate host defense and pathological inflammation. We have previously reported that host dendritic cells (DCs) infected with Vibrio vulnificus induce Th17 responses through the production of several pro-inflammatory cytokines, including interleukin (IL)-1ß and IL-6. V. vulnificus produces RTX toxin (RtxA), an important virulence factor that determines successful pathophysiology. In this study, we investigated the involvement of RtxA from V. vulnificus in Th17 cell induction through the activation and maturation of DCs. The increased expression of the DC surface marker CD40 caused by V. vulnificus wild-type infection was reduced by rtxA gene mutation in V. vulnificus. The mRNA and protein levels of Th17 polarization-related cytokines also decreased in V. vulnificus rtxA mutant-infected DCs. In addition, the co-culture of Th cells and DCs infected with rtxA mutant V. vulnificus resulted in reduction in DC-mediated Th17 responses. Th17 cell responses in the small intestinal lamina propria decreased in mice inoculated with V. vulnificus rtxA mutant as compared to those inoculated with the wild-type strain. These decreases in DC maturation, Th17-polarizing cytokine secretion, and Th17 responses attributed to rtxA mutation were restored following infection with the rtxA revertant strain. Furthermore, the mutation in the hlyU gene encoding the activator of rtxA1 gene reproduced the results observed with rtxA mutation. Taken together, V. vulnificus, by means of RtxA, induces inflammatory Th17 responses, which may be associated with adaptive responses of the host against V. vulnificus infection.


Asunto(s)
Toxinas Bacterianas/inmunología , Inflamación/inmunología , Células Th17/inmunología , Vibriosis/inmunología , Vibrio vulnificus/inmunología , Animales , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Células Th17/metabolismo , Células Th17/microbiología , Vibriosis/metabolismo , Vibriosis/microbiología , Vibrio vulnificus/fisiología
12.
mBio ; 9(1)2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382732

RESUMEN

Pathogenic Vibrio species cause diseases in diverse marine animals reared in aquaculture. Since their pathogenesis, persistence, and survival in marine environments are regulated by quorum sensing (QS), QS interference has attracted attention as a means to control these bacteria in aquatic settings. A few QS inhibitors of Vibrio species have been reported, but detailed molecular mechanisms are lacking. Here, we identified a novel, potent, and selective Vibrio QS inhibitor, named QStatin [1-(5-bromothiophene-2-sulfonyl)-1H-pyrazole], which affects Vibrio harveyi LuxR homologues, the well-conserved master transcriptional regulators for QS in Vibrio species. Crystallographic and biochemical analyses showed that QStatin binds tightly to a putative ligand-binding pocket in SmcR, the LuxR homologue in V. vulnificus, and changes the flexibility of the protein, thereby altering its transcription regulatory activity. Transcriptome analysis revealed that QStatin results in SmcR dysfunction, affecting the expression of SmcR regulon required for virulence, motility/chemotaxis, and biofilm dynamics. Notably, QStatin attenuated representative QS-regulated phenotypes in various Vibrio species, including virulence against the brine shrimp (Artemia franciscana). Together, these results provide molecular insights into the mechanism of action of an effective, sustainable QS inhibitor that is less susceptible to resistance than other antimicrobial agents and useful in controlling the virulence of Vibrio species in aquacultures.IMPORTANCE Yields of aquaculture, such as penaeid shrimp hatcheries, are greatly affected by vibriosis, a disease caused by pathogenic Vibrio infections. Since bacterial cell-to-cell communication, known as quorum sensing (QS), regulates pathogenesis of Vibrio species in marine environments, QS inhibitors have attracted attention as alternatives to conventional antibiotics in aquatic settings. Here, we used target-based high-throughput screening to identify QStatin, a potent and selective inhibitor of V. harveyi LuxR homologues, which are well-conserved master QS regulators in Vibrio species. Structural and biochemical analyses revealed that QStatin binds tightly to a putative ligand-binding pocket on SmcR, the LuxR homologue in V. vulnificus, and affects expression of QS-regulated genes. Remarkably, QStatin attenuated diverse QS-regulated phenotypes in various Vibrio species, including pathogenesis against brine shrimp, with no impact on bacterial viability. Taken together, the results suggest that QStatin may be a sustainable antivibriosis agent useful in aquacultures.


Asunto(s)
Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Percepción de Quorum/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Vibrio/efectos de los fármacos , Cristalografía por Rayos X , Perfilación de la Expresión Génica , Unión Proteica , Proteínas Represoras/química , Transactivadores/química
13.
IUCrJ ; 5(Pt 1): 82-92, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29354274

RESUMEN

Peroxiredoxins (Prxs) are ubiquitous cysteine-based peroxidase enzymes. Recently, a new type of Prx, VvPrx3, was identified in the pathogenic bacterium Vibrio vulnificus as being important for survival in macrophages. It employs only one catalytic cysteine residue to decompose peroxides. Here, crystal structures of VvPrx3 representing its reduced and oxidized states have been determined, together with an H2O2-bound structure, at high resolution. The crystal structure representing the reduced Prx3 showed a typical dimeric interface, called the A-type interface. However, VvPrx3 forms an oligomeric interface mediated by a disulfide bond between two catalytic cysteine residues from two adjacent dimers, which differs from the doughnut-like oligomers that appear in most Prxs. Subsequent biochemical studies showed that this disulfide bond was induced by treatment with nitric oxide (NO) as well as with peroxides. Consistently, NO treatment induced expression of the prx3 gene in V. vulnificus, and VvPrx3 was crucial for the survival of bacteria in the presence of NO. Taken together, the function and mechanism of VvPrx3 in scavenging peroxides and NO stress via oligomerization are proposed. These findings contribute to the understanding of the diverse functions of Prxs during pathogenic processes at the molecular level.

14.
Int J Mol Med ; 41(1): 531-540, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115398

RESUMEN

Vibrio vulnificus (V. vulnificus) is a gram-negative bacterium, which causes life-threatening septicemia and gastroenteritis through the consumption of contaminated seafood or wound infection. In addition, V. vulnificus infection is known to stimulate the production of several pro-inflammatory cytokines, which are associated with inflammatory responses mediated predominantly by dendritic cells (DCs), functioning as antigen-presenting cells. The present study aimed to investigate whether V. vulnificus infection induced the maturation and activation of murine DCs, which have the ability to polarize T helper (Th) cells into Th17 cells. Dysregulated Th17 cell responses are known to cause tissue damage, promoting the penetration of pathogens; however, Th17 cells are also involved in host defense against infection. Infection with V. vulnificus significantly increased the expression of cell surface molecules, including CD40, CD80 and major histocompatibility complex class II, leading to the maturation and activation of DCs. In the present study, the analysis of the cytokine profiles of DCs upon infection with V. vulnificus revealed the preferential production of interleukin-1ß (IL-1ß) and IL-6, through which V. vulnificus-infected DCs induced the polarization of Th17 cells when naïve CD4+ T cells were co-incubated. The reduction of Th17 cell generation through the use of anti-IL-6 neutralizing antibodies indicated that the Th17-polarizing capacity of V. vulnificus was predominantly dependent on DC-derived IL-6. The in vivo administration of V. vulnificus-infected DCs consistently increased the Th17 cell population in the lymph nodes of mice. Finally, the oral administration of V. vulnificus in mice also increased Th17 cell responses in the lamina propria of the small intestine. These results collectively demonstrated that V. vulnificus induced inflammatory Th17 cell responses via DCs, which may be associated with the immunopathological effects caused by V. vulnificus infection.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Inflamación/inmunología , Células Th17/inmunología , Vibriosis/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Antígeno B7-1/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Polaridad Celular/inmunología , Humanos , Inflamación/genética , Inflamación/microbiología , Inflamación/patología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Ganglios Linfáticos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Vibriosis/genética , Vibriosis/microbiología , Vibriosis/patología , Vibrio vulnificus/inmunología , Vibrio vulnificus/patogenicidad
15.
J Biol Chem ; 292(41): 17129-17143, 2017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-28855258

RESUMEN

The marine bacterium Vibrio vulnificus causes food-borne diseases, which may lead to life-threatening septicemia in some individuals. Therefore, identifying virulence factors in V. vulnificus is of high priority. We performed a transcriptome analysis on V. vulnificus after infection of human intestinal HT29-methotrexate cells and found induction of plpA, encoding a putative phospholipase, VvPlpA. Bioinformatics, biochemical, and genetic analyses demonstrated that VvPlpA is a phospholipase A2 secreted in a type II secretion system-dependent manner. Compared with the wild type, the plpA mutant exhibited reduced mortality, systemic infection, and inflammation in mice as well as low cytotoxicity toward the human epithelial INT-407 cells. Moreover, plpA mutation attenuated the release of actin and cytosolic cyclophilin A from INT-407 cells, indicating that VvPlpA is a virulence factor essential for causing lysis and necrotic death of the epithelial cells. plpA transcription was growth phase-dependent, reaching maximum levels during the early stationary phase. Also, transcription factor HlyU and cAMP receptor protein (CRP) mediate additive activation and host-dependent induction of plpA Molecular biological analyses revealed that plpA expression is controlled via the promoter, P plpA , and that HlyU and CRP directly bind to P plpA upstream sequences. Taken together, this study demonstrated that VvPlpA is a type II secretion system-dependent secretory phospholipase A2 regulated by HlyU and CRP and is essential for the pathogenicity of V. vulnificus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fosfolipasas A2/metabolismo , Vibriosis/enzimología , Vibrio vulnificus/enzimología , Vibrio vulnificus/patogenicidad , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Línea Celular , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Fosfolipasas A2/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vibriosis/genética , Vibriosis/patología , Vibrio vulnificus/genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-28848713

RESUMEN

An inflammatory form of phagocyte death evoked by the Gram-negative bacterium Vibrio (V.) vulnificus (WT) is one of hallmarks to promote their colonization, but the virulence factor and infectious mechanism involved in this process remain largely unknown. Here, we identified extracellular metalloprotease VvpM as a new virulence factor and investigated the molecular mechanism of VvpM which acts during the regulation of the inflammatory form of macrophage death and bacterial colonization. Mutation of the vvpM gene appeared to play major role in the prevention of IL-1ß production due to V. vulnificus infection in macrophage. However, the recombinant protein (r) VvpM caused IL-1ß production coupled with necrotic cell death, which is highly susceptible to the knockdown of annexin A2 (ANXA2) located in both membrane lipid and non-lipid rafts. In lipid rafts, rVvpM recruited NOX enzymes coupled with ANXA2 to facilitate the production of ROS responsible for the epigenetic and transcriptional regulation of NF-κB in the IL-1ß promoter. rVvpM acting on non-lipid rafts increased LC3 puncta formation and autophagic flux, which are required for the mRNA expression of Atg5 involved in the autophagosome formation process. The autophagy activation caused by rVvpM induced NLRP3 inflammasome-dependent caspase-1 activation in the promoting of IL-1ß production. In mouse models of V. vulnificus infection, the VvpM mutant failed to elevate the level of pro-inflammatory responses closely related to IL-1ß production and prevented bacterial colonization. These findings delineate VvpM efficiently regulates two pathogenic pathways that stimulate NF-κB-dependent IL-1ß production and autophagy-mediated NLRP3 inflammasome via distinct spatial targeting by ANXA2.


Asunto(s)
Anexina A2/metabolismo , Apoptosis , Interleucina-1beta/metabolismo , Macrófagos/fisiología , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Anexina A2/genética , Células CACO-2 , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/genética , Microdominios de Membrana/metabolismo , Metaloproteasas/genética , Metaloproteasas/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vibriosis/patología , Factores de Virulencia/genética
17.
Mol Cells ; 40(4): 299-306, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28427249

RESUMEN

The transcriptional activator AphB has been implicated in acid resistance and pathogenesis in the food borne pathogens Vibrio vulnificus and Vibrio cholerae. To date, the full-length AphB crystal structure of V. cholerae has been determined and characterized by a tetrameric assembly of AphB consisting of a DNA binding domain and a regulatory domain (RD). Although acidic pH and low oxygen tension might be involved in the activation of AphB, it remains unknown which ligand or stimulus activates AphB at the molecular level. In this study, we determine the crystal structure of the AphB RD from V. vulnificus under aerobic conditions without modification at the conserved cysteine residue of the RD, even in the presence of the oxidizing agent cumene hydroperoxide. A cysteine to serine amino acid residue mutant RD protein further confirmed that the cysteine residue is not involved in sensing oxidative stress in vitro. Interestingly, an unidentified small molecule was observed in the inter-subdomain cavity in the RD when the crystal was incubated with cumene hydroperoxide molecules, suggesting a new ligand-binding site. In addition, we confirmed the role of AphB in acid tolerance by observing an aphB-dependent increase in cadC transcript level when V. vulnificus was exposed to acidic pH. Our study contributes to the understanding of the AphB molecular mechanism in the process of recognizing the host environment.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/fisiología , Genes Reguladores , Transactivadores/química , Transactivadores/fisiología , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Derivados del Benceno/química , Sitios de Unión , Cristalografía por Rayos X , Cisteína/química , Cisteína/genética , Regulación Bacteriana de la Expresión Génica , Concentración de Iones de Hidrógeno , Ligandos , Modelos Moleculares , Estrés Oxidativo/fisiología , Dominios y Motivos de Interacción de Proteínas , Análisis de Secuencia de Proteína , Serina/química , Serina/genética , Vibrio vulnificus/genética , Virulencia/genética
18.
Sci Rep ; 6: 27080, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27250250

RESUMEN

VvhA, a virulent factor of Vibrio (V.) vulnificus, induces acute cell death in a destructive manner. Autophagy plays an important role in cell death, but the functional role of VvhA in autophagy-related cell death has not been elucidated yet. We found that rVvhA significantly increased LC3 puncta formation and autophagic flux in promoting the cell death of human intestinal epithelial Caco-2 cells. The cell death induced by rVvhA was independent of lysosomal permeabilizaton and caspase activation. rVvhA induced rapid phosphorylation of c-Src in the membrane lipid raft, which resulted in an increased interaction between lipid raft molecule caveolin-1 and NADPH oxidase (NOX) complex Rac1 for ROS production. NOX-mediated ROS signaling induced by rVvhA increased the phosphorylation of extracellular signal-regulated kinase (ERK) and eukaryotic translation initiation factor 2α (eIF2α) which are required for mRNA expression of Atg5 and Atg16L1 involved in autophagosome formation. In an in vivo model, VvhA increased autophagy activation and paracellular permeabilization in intestinal epithelium. Collectively, the results here show that VvhA plays a pivotal role in the pathogenesis and dissemination of V. vulnificus by autophagy upregulation, through the lipid raft-mediated c-Src/NOX signaling pathway and ERK/eIF2α activation.


Asunto(s)
Autofagia , Microdominios de Membrana/metabolismo , Transducción de Señal , Vibriosis/patología , Vibrio vulnificus/fisiología , Animales , Proteínas Bacterianas/fisiología , Proteína Tirosina Quinasa CSK , Células CACO-2 , Caspasas/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Intestinos/patología , Lisosomas/metabolismo , Microdominios de Membrana/microbiología , Ratones Endogámicos ICR , NADPH Oxidasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Vibriosis/microbiología , Familia-src Quinasas/metabolismo
19.
J Biol Chem ; 291(11): 5774-5787, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26755724

RESUMEN

Binding to mucin is the initial step for enteropathogens to establish pathogenesis. An open reading frame, gbpA, of Vibrio vulnificus was identified and characterized in this study. Compared with wild type, the gbpA mutant was impaired in binding to mucin-agar and the mucin-secreting HT29-methotrexate cells, and the impaired mucin binding was restored by the purified GbpA provided exogenously. The gbpA mutant had attenuated virulence and ability of intestinal colonization in a mouse model, indicating that GbpA is a mucin-binding protein and essential for pathogenesis of V. vulnificus. The gbpA transcription was growth phase-dependent, reaching a maximum during the exponential phase. The Fe-S cluster regulator (IscR) and the cyclic AMP receptor protein (CRP) coactivated, whereas SmcR, a LuxR homologue, repressed gbpA. The cellular levels of IscR, CRP, and SmcR were not significantly affected by one another, indicating that the regulator proteins function cooperatively to regulate gbpA rather than sequentially in a regulatory cascade. The regulatory proteins directly bind upstream of the gbpA promoter PgbpA. DNase I protection assays, together with the deletion analyses of PgbpA, demonstrated that IscR binds to two specific sequences centered at -164.5 and -106, and CRP and SmcR bind specifically to the sequences centered at -68 and -45, respectively. Furthermore, gbpA was induced by exposure to H2O2, and the induction appeared to be mediated by elevated intracellular levels of IscR. Consequently, the combined results indicated that IscR, CRP, and SmcR cooperate for precise regulation of gbpA during the V. vulnificus pathogenesis.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mucinas/metabolismo , Vibriosis/metabolismo , Vibriosis/patología , Vibrio vulnificus/genética , Vibrio vulnificus/fisiología , Animales , Secuencia de Bases , Femenino , Regulación Bacteriana de la Expresión Génica , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Mutación , Estrés Oxidativo , Unión Proteica , Percepción de Quorum , Sitio de Iniciación de la Transcripción , Vibriosis/microbiología
20.
Int J Med Microbiol ; 306(1): 10-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26552364

RESUMEN

The disruption of gastrointestinal tight junctions and their colonization evoked by enteric pathogens are hallmarks of the pathogenesis. Vibrio (V.) vulnificus, VvpE, is an elastase which is responsible for host surface adherence and vascular permeability; however, the functional roles of VvpE in the pathogenesis of V. vulnificus (WT) are poorly understood. In the present study, we have investigated the role of VvpE in regulation of intestinal tight junctions and the colonization of WT. We found that mutation of the vvpE gene from V. vulnificus (vvpE mutant) prevents intestinal tight/adherens junction dysregulation due to a WT infection and maintains the physiological level of the epithelial paracellular permeability. Interestingly, the vvpE mutant exhibited defective intestinal colonization abilities, whereas WT colonization was significantly elevated in the ileum in a time-dependent manner. Finally, the vvpE mutant negated the enterotoxicity, the breakdown of red blood cells, and pro-inflammatory responses, all of which are induced by the WT infection. In addition, the results of a LC-MS/MS analysis showed that VvpE contributes to WT pathogenesis in multiple ways by interacting with intestinal proteins, including ß-globin, Annexin A2, Annexin A4, F-actin, and intelectin-1b. These results demonstrate that VvpE plays important role in promoting the tight junction disruption and intestinal colonization of V. vulnificus and that it also has the ability to interact with the intestinal proteins responsible for microbial pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/efectos de los fármacos , Metaloendopeptidasas/metabolismo , Elastasa Pancreática/metabolismo , Uniones Estrechas/efectos de los fármacos , Vibrio vulnificus/fisiología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Células Epiteliales/fisiología , Técnicas de Inactivación de Genes , Masculino , Metaloendopeptidasas/genética , Ratones Endogámicos ICR , Elastasa Pancreática/genética , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...