Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Commun Signal ; 11: 79, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24144181

RESUMEN

BACKGROUND: Enteric pathogens utilize a distinct set of proteins to modulate host cell signaling events that promote host cell invasion, induction of the inflammatory response, and intracellular survival. Human infection with Campylobacter jejuni, the causative agent of campylobacteriosis, is characterized by diarrhea containing blood and leukocytes. The clinical presentation of acute disease, which is consistent with cellular invasion, requires the delivery of the Campylobacter invasion antigens (Cia) to the cytosol of host cells via a flagellar Type III Secretion System (T3SS). We identified a novel T3SS effector protein, which we termed CiaD that is exported from the C. jejuni flagellum and delivered to the cytosol of host cells. RESULTS: We show that the host cell kinases p38 and Erk 1/2 are activated by CiaD, resulting in the secretion of interleukin-8 (IL-8) from host cells. Additional experiments revealed that CiaD-mediated activation of p38 and Erk 1/2 are required for maximal invasion of host cells by C. jejuni. CiaD contributes to disease, as evidenced by infection of IL-10 knockout mice. Noteworthy is that CiaD contains a Mitogen-activated protein (MAP) kinase-docking site that is found within effector proteins produced by other enteric pathogens. These findings indicate that C. jejuni activates the MAP kinase signaling pathways Erk 1/2 and p38 to promote cellular invasion and the release of the IL-8 pro-inflammatory chemokine. CONCLUSIONS: The identification of a novel T3SS effector protein from C. jejuni significantly expands the knowledge of virulence proteins associated with C. jejuni pathogenesis and provides greater insight into the mechanism utilized by C. jejuni to invade host cells.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Campylobacter/metabolismo , Campylobacter jejuni/fisiología , Sistema de Señalización de MAP Quinasas , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Sitios de Unión , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/patogenicidad , Línea Celular , Flagelos/metabolismo , Humanos , Interleucina-10/genética , Interleucina-8/metabolismo , Ratones , Ratones Noqueados , Mutación , Factores de Virulencia/genética
2.
Microorganisms ; 11(1)2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36677486

RESUMEN

Many antibiotic-resistant bacteria carry resistance genes on conjugative plasmids that are transferable to commensals and pathogens. We determined the ability of multiple enteric bacteria to acquire and retransfer a broad-host-range plasmid RP4. We used human-derived commensal Escherichia coli LM715-1 carrying a chromosomal red fluorescent protein gene and green fluorescent protein (GFP)-labeled broad-host-range RP4 plasmid with ampR, tetR, and kanR in in vitro matings to rifampicin-resistant recipients, including Escherichia coli MG1655, Dec5α, Vibrio cholerae, Pseudomonas putida, Pseudomonas aeruginosa, Klebsiella pneumoniae, Citrobacter rodentium, and Salmonella Typhimurium. Transconjugants were quantified on selective media and confirmed using fluorescence microscopy and PCR for the GFP gene. The plasmid was transferred from E. coli LM715-1 to all tested recipients except P. aeruginosa. Transfer frequencies differed between specific donor-recipient pairings (10-2 to 10-8). Secondary retransfer of plasmid from transconjugants to E. coli LM715-1 occurred at frequencies from 10-2 to 10-7. A serial passage plasmid persistence assay showed plasmid loss over time in the absence of antibiotics, indicating that the plasmid imposed a fitness cost to its host, although some plasmid-bearing cells persisted for at least ten transfers. Thus, the RP4 plasmid can transfer to multiple clinically relevant bacterial species without antibiotic selection pressure.

3.
bioRxiv ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961091

RESUMEN

The mammalian gut microbiome influences numerous developmental processes. In human infants it has been linked with cognition, social skills, hormonal responses to stress, and brain connectivity. Yet, these associations are not necessarily causal. The present study tested whether two microbial stool communities, common in human infants, affected behavior, myelination, dendritic morphology, and spine density when used to colonize mouse models. Humanized animals were more like specific-pathogen free mice than germ-free mice for most phenotypes, although in males, both humanized groups were less social. Both humanized groups had thinner myelin sheaths in the hippocampus, than did germ-free animals. Humanized animals were similar to each other except for dendritic morphology and spine density where one group had greater dendritic length in the prefrontal cortex, greater dendritic volume in the nucleus accumbens, and greater spine density in both regions, compared to the other. Results add to a body of literature suggesting the gut microbiome impacts brain development. Teaser: Fecal transplants from human infants with highly abundant Bifidobacterium , an important inhabitant of the intestinal tract of breastfed newborns, may promote brain connectivity in mice.

4.
J Bacteriol ; 194(20): 5707-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23012285

RESUMEN

The Campylobacter jejuni human clinical isolates NW and D2600 colonized C57BL/6 interleukin-10-deficient (IL-10(-/-)) mice without inducing a robust inflammatory response (J. A. Bell et al., BMC Microbiol. 9:57, 2009). We announce draft genome sequences of NW and D2600 to facilitate comparisons with strains that induce gastrointestinal inflammation in this mouse model.


Asunto(s)
Campylobacter jejuni/genética , ADN Bacteriano/química , ADN Bacteriano/genética , Genoma Bacteriano , Análisis de Secuencia de ADN , Animales , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/aislamiento & purificación , Modelos Animales de Enfermedad , Humanos , Interleucina-10/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular
5.
Microbiology (Reading) ; 158(Pt 5): 1304-1316, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22343355

RESUMEN

Human illness due to Camplyobacter jejuni infection is closely associated with consumption of poultry products. We previously demonstrated a 50 % shift in allele frequency (phase variation) in contingency gene Cj1139 (wlaN) during passage of C. jejuni NCTC11168 populations through Ross 308 broiler chickens. We hypothesized that phase variation in contingency genes during chicken passage could promote subsequent colonization and disease in humans. To test this hypothesis, we passaged C. jejuni strains NCTC11168, 33292, 81-176, KanR4 and CamR2 through broiler chickens and analysed the ability of passaged and non-passaged populations to colonize C57BL6 IL-10-deficient mice, our model for human colonization and disease. We utilized fragment analysis and nucleotide sequence analysis to measure phase variation in contingency genes. Passage through the chicken reservoir promoted phase variation in five specific contingency genes, and these 'successful' populations colonized mice. When phase variation did not occur in these same five contingency genes during chicken passage, these 'unsuccessful' populations failed to colonize mice. Phase variation during chicken passage generated small insertions or deletions (indels) in the homopolymeric tract (HT) in contingency genes. Single-colony isolates of C. jejuni strain KanR4 carrying an allele of contingency gene Cj0170 with a10G HT colonized mice at high frequency and caused disease symptoms, whereas single-colony isolates carrying the 9G allele failed to colonize mice. Supporting results were observed for the successful 9G allele of Cj0045 in strain 33292. These data suggest that phase variation in Cj0170 and Cj0045 is strongly associated with mouse colonization and disease, and that the chicken reservoir can play an active role in natural selection, phase variation and disease.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/genética , Campylobacter jejuni/patogenicidad , Factores de Virulencia/genética , Alelos , Animales , Campylobacter jejuni/crecimiento & desarrollo , Pollos/microbiología , Regulación Bacteriana de la Expresión Génica , Frecuencia de los Genes , Mutación INDEL , Ratones , Ratones Endogámicos C57BL , Pase Seriado , Virulencia
6.
Gut Microbes ; 14(1): 2064706, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35442154

RESUMEN

Campylobacter jejuni is a leading cause of gastroenteritis that has been causally linked with development of the autoimmune peripheral neuropathy Guillain Barré Syndrome (GBS). Previously, we showed that C. jejuni isolates from human enteritis patients induced Type1/17-cytokine dependent colitis in interleukin-10 (IL-10)-/- mice, while isolates from GBS patients colonized these mice without colitis but instead induced autoantibodies that cross-reacted with the sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni and the peripheral nerve gangliosides. We show here that infection of IL-10-/- mice with the GBS but not the colitis isolate led to sciatic nerve inflammation and abnormal gait and hind limb movements, with character and timing consistent with this syndrome in humans. Autoantibody responses and associated nerve histologic changes were dependent on IL-4 production by CD4 T cells. We further show that Siglec-1 served as a central antigen presenting cell receptor mediating the uptake of the GBS isolates via interaction with the sialylated oligosaccharide motifs found specifically on the LOS of GBS-associated C. jejuni, and the ensuing T cell differentiation and autoantibody elicitation. Sialylated oligosaccharide motifs on the LOS of GBS-associated C. jejuni therefore acted as both the Siglec-1-ligand for phagocytosis, as well as the epitope for autoimmunity. Overall, we present a mouse model of an autoimmune disease induced directly by a bacterium that is dependent upon Siglec-1 and IL-4. We also demonstrate the negative regulatory role of IL-10 in C. jejuni induced autoimmunity and provide IL-4 and Siglec-1 blockade as potential therapeutic interventions against GBS.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Colitis , Microbioma Gastrointestinal , Síndrome de Guillain-Barré , Animales , Autoanticuerpos , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/genética , Colitis/microbiología , Síndrome de Guillain-Barré/etiología , Síndrome de Guillain-Barré/patología , Humanos , Interleucina-10/genética , Interleucina-4 , Lipopolisacáridos , Ratones , Lectina 1 Similar a Ig de Unión al Ácido Siálico
7.
Comp Med ; 72(2): 63-77, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35272743

RESUMEN

Campylobacter jejuni is an important cause of bacterial gastroenteritis worldwide and is linked to Guillain-Barré syndrome (GBS), a debilitating postinfectious polyneuropathy. The immunopathogenesis of GBS involves the generation of antibodies that are cross reactive to C. jejuni lipooligosaccharide and structurally similar peripheral nerve gangliosides. Both the C. jejuni infecting strain and host factors contribute to GBS development. GBS pathogenesis is associated with Th2-mediated responses in patients. Moreover, induction of IgG1 antiganglioside antibodies in association with colonic Th2-mediated immune responses has been reported in C. jejuni-infected C57BL/6 IL10-/- mice at 4 to 6 wk after infection. We hypothesized that, due to their Th2 immunologic bias, BALB/c mice would develop autoantibodies and signs of peripheral neuropathy after infection with a GBS patient-derived strain of C. jejuni (strain 260.94). WT and IL10-/- BALB/c mice were orally inoculated with C. jejuni 260.94, phenotyped weekly for neurologic deficits, and euthanized after 5 wk. Immune responses were assessed as C. jejuni-specific and antiganglioside antibodies in plasma and cytokine production and histologic lesions in the proximal colon. Peripheral nerve lesions were assessed in dorsal root ganglia and their afferent nerve fibers by scoring immunohistochemically labeled macrophages through morphometry. C. jejuni 260.94 stably colonized both WT and IL10-/- mice and induced systemic Th1/Th17-mediated immune responses with significant increases in C. jejuni-specific IgG2a, IgG2b, and IgG3 plasma antibodies. However, C. jejuni 260.94 did not induce IgG1 antiganglioside antibodies, colitis, or neurologic deficits or peripheral nerve lesions in WT or IL10-/- mice. Both WT and IL10-/- BALB/c mice showed relative protection from development of Th2-mediated immunity and antiganglioside antibodies as compared with C57BL/6 IL10-/- mice. Therefore, BALB/c mice infected with C. jejuni 260.94 are not an effective disease model but provide the opportunity to study the role of immune mechanisms and host genetic background in the susceptibility to post infectious GBS.


Asunto(s)
Infecciones por Campylobacter , Campylobacter jejuni , Síndrome de Guillain-Barré , Animales , Infecciones por Campylobacter/complicaciones , Humanos , Inmunoglobulina G , Interleucina-10 , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
8.
Front Microbiol ; 13: 800269, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35591997

RESUMEN

Campylobacter jejuni causes foodborne gastroenteritis and may trigger acute autoimmune sequelae including Guillain Barré Syndrome. Onset of neuromuscular paralysis is associated with exposure to C. jejuni lipooligosaccharide (LOS) classes A, B, C, D, and E that mimic and evoke antibodies against gangliosides on myelin and axons of peripheral nerves. Family members managing a Michigan dairy operation reported recurring C. jejuni gastroenteritis. Because dairy cattle are known to shed C. jejuni, we hypothesized that calves in the sick pen were the source of human infections. Fecal samples obtained from twenty-five calves, one dog, and one asymptomatic family member were cultured for Campylobacter. C. jejuni isolates were obtained from thirteen calves and the family member: C. coli from two calves, and C. hyointestinalis from two calves. Some calves had diarrhea; most were clinically normal. Typing of lipooligosaccharide biosynthetic loci showed that eight calf C. jejuni isolates fell into classes A, B, and C. Two calf isolates and the human isolate possessed LOS class E, associated mainly with enteric disease and rarely with Guillain Barré Syndrome. Multi-locus sequence typing, porA and flaA typing, and whole genome comparisons of the thirteen C. jejuni isolates indicated that the three LOS class E strains that included the human isolate were closely related, indicating zoonotic transmission. Whole-genome comparisons revealed that isolates differed in virulence gene content, particularly in loci encoding biosynthesis of surface structures. Family members experienced diarrheal illness repeatedly over 2 years, yet none experienced GBS despite exposure to calves carrying invasive C. jejuni with LOS known to elicit antiganglioside autoantibodies.

9.
Comp Med ; 71(1): 46-65, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33334395

RESUMEN

The nematode Trichuris muris has been shown to interact with specific enteric bacteria, but its effects on the composition of its host's microbial community are not fully understood. We hypothesized that Trichuris muris-infected mice would have altered colon microbiota as compared with uninfected mice. Colon histopathology and microbial community structure and composition were examined in mouse models of colitis (C3BirTLR4-/- IL10-/- and C3H/HeJ TLR4-/- IL10+/+ mice) with and without T. muris infection, in uninfected C3BirIL10-/- mice with and without spontaneous colitis, and in normal C3H/ HeJ mice. T. muris-infected mice developed colon lesions that were more severe than those seen in IL10-deficient mice. Ap- proximately 80% of infected IL10-/- mice had colon neutrophilic exudates, and some had extraintestinal worms and bacteria. The composition and structure of proximal colon microbiota were assessed by using terminal restriction fragment length polymorphism analysis targeting the bacterial 16S rRNA gene. Colon microbiota in C3BirIL10-/- and C3H/HeJ mice differed both qualitatively and quantitatively. Trichuris infection significantly altered the relative abundance of individual operational taxonomic units [OTU] but not the composition (presence or absence of OTU) of colon microbiota in the 2 mouse genotypes. When C3BirIL10-/- and C3H/HeJ mouse OTU were considered separately, Trichuris was found to affect the microbiota of C3BirIL10-/- mice but not of C3H/HeJ mice. Even though 34 of the 75 (45%) C3BirIL10-/- mice had spontaneous colitis, neither qualitative nor quantitative differences were detected in microbiota between colitic or noncolitic C3BirIL10-/- mice or noncolitic C3H/HeJ mice. Therefore, Trichuris-infected mice developed distinct microbial communities that were influenced by host background genes; these alterations cannot be attributed solely to colonic inflammation.


Asunto(s)
Colitis , Microbiota , Animales , Interleucina-10/genética , Ratones , Ratones Endogámicos C3H , ARN Ribosómico 16S , Trichuris
10.
Microbiology (Reading) ; 156(Pt 7): 2046-2057, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20360176

RESUMEN

Previous studies have demonstrated that Campylobacter jejuni, the leading causative agent of bacterial food-borne disease in the USA, exhibits high-frequency genetic variation that is associated with changes in cell-surface antigens and ability to colonize chickens. To expand our understanding of the role of genetic diversity in the disease process, we analysed the ability of three C. jejuni human disease isolates (strains 11168, 33292 and 81-176) and genetically marked derivatives to colonize Ross 308 broilers and C57BL/6J IL10-deficient mice. C. jejuni colonized broilers at much higher efficiency (all three strains, 23 of 24 broilers) than mice (11168 only, 8 of 24 mice). C. jejuni 11168 genetically marked strains colonized mice at very low efficiency (2 of 42 mice); however, C. jejuni reisolated from mice colonized both mice and broilers at high efficiency, suggesting that this pathogen can adapt genetically in the mouse. We compared the genome composition in the three wild-type C. jejuni strains and derivatives by microarray DNA/DNA hybridization analysis; the data demonstrated a high degree of genetic diversity in three gene clusters associated with synthesis and modification of the cell-surface structures capsule, flagella and lipo-oligosaccharide. Finally, we analysed the frequency of mutation in homopolymeric tracts associated with the contingency genes wlaN (GC tract) and flgR (AT tracts) in culture and after passage through broilers and mice. C. jejuni adapted genetically in culture at high frequency and the degree of genetic diversity was increased by passage through broilers but was nearly eliminated in the gastrointestinal tract of mice. The data suggest that the broiler gastrointestinal tract provides an environment which promotes outgrowth and genetic variation in C. jejuni; the enhancement of genetic diversity at this location may contribute to its importance as a human disease reservoir.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/genética , Pollos/microbiología , Reservorios de Enfermedades/microbiología , Variación Genética , Ratones/microbiología , Animales , Proteínas Bacterianas/genética , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Parasitol Res ; 107(5): 1103-13, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20809420

RESUMEN

Murine immunological responses to experimental infection with Trichuris muris and the effects of the resident microbiota on these responses are of increasing interest. For these studies, accurate dose delivery and improved sterilization of inocula are essential to prevent co-infection with unknown contaminants. We found that washing T. muris eggs with antibiotics may not be sufficient for sterilization of inocula. However, washing eggs in 6.25% hypochlorite/bleach eliminated bacteria and fungi, as determined by culture and PCR, did not harm viable T. muris eggs and reduced the number of non-viable eggs in the inocula. A hatching assay and propidium iodide staining method were developed and found to increase the accuracy for assessing T. muris egg viability prior to infection for rapid dose evaluation. In addition, metal gavage feeding needles increased the accuracy and precision of the dose delivered to the mice compared to flexible rubber tubes. These methods will improve experimental Trichuris studies by decreasing the variability in outcome due to unintended carryover of adherent microorganisms and unrecognized variation in inocula.


Asunto(s)
Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos , Tricuriasis/inmunología , Tricuriasis/parasitología , Trichuris/inmunología , Trichuris/patogenicidad , Animales , Supervivencia Celular , Descontaminación/métodos , Colorantes Fluorescentes/farmacología , Ratones , Parasitología/métodos , Propidio/farmacología , Coloración y Etiquetado/métodos , Trichuris/crecimiento & desarrollo
12.
Front Microbiol ; 11: 579989, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240235

RESUMEN

Evolution experiments in the laboratory have focused heavily on model organisms, often to the exclusion of clinically relevant pathogens. The foodborne bacterial pathogen Campylobacter jejuni belongs to a genus whose genomes are small compared to those of its closest genomic relative, the free-living genus Sulfurospirillum, suggesting genome reduction during the course of evolution to host association. In an in vitro experiment, C. jejuni serially passaged in rich medium in the laboratory exhibited loss of flagellar motility-an essential function for host colonization. At early time points the motility defect was often reversible, but after 35 days of serial culture, motility was irreversibly lost in most cells in 5 independently evolved populations. Population re-sequencing revealed disruptive mutations to genes in the flagellar transcriptional cascade, rpoN (σ54)-therefore disrupting the expression of the genes σ54 regulates-coupled with deletion of rpoN in all evolved lines. Additional mutations were detected in virulence-related loci. In separate in vivo experiments, we demonstrate that a phase variable (reversible) motility mutant carrying an adenine deletion within a homopolymeric tract resulting in truncation of the flagellar biosynthesis gene fliR was deficient for colonization in a C57BL/6 IL-10-/- mouse disease model. Re-insertion of an adenine residue partially restored motility and ability to colonize mice. Thus, a pathogenic C. jejuni strain was rapidly attenuated by experimental laboratory evolution and demonstrated genomic instability during this evolutionary process. The changes observed suggest C. jejuni is able to evolve in a novel environment through genome reduction as well as transition, transversion, and slip-strand mutations.

13.
Infect Immun ; 77(6): 2499-507, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19332531

RESUMEN

Campylobacter jejuni is an important cause of human enteritis and has been linked to the development of autoimmune diseases. Recently we showed that infection of murine dendritic cells (DCs) with C. jejuni resulted in DC activation and induction of Campylobacter-specific Th1-effector responses. Toll-like receptor (TLR) signaling through myeloid differentiation factor 88 (MyD88) and/or Toll-interleukin 1 (IL-1) receptor domain-containing adaptor-inducing beta interferon (IFN-beta) (TRIF) is critical in inducing immunity against pathogens. In this study, we investigated the role of TLR2, TLR4, MyD88, and TRIF signaling in C. jejuni-induced inflammatory activation of DCs. DC upregulation of major histocompatibility complex class II and costimulatory molecules after C. jejuni challenge was profoundly impaired by TLR2, TLR4, MyD88, and TRIF deficiencies. Similarly, C. jejuni-induced secretion of IL-12, IL-6, and tumor necrosis factor alpha was significantly inhibited in TLR2(-/-), TLR4(-/-), MyD88(-/-), and TRIF(-/-) DCs compared to that in wild-type DCs; however, the magnitude of inhibition was greater in MyD88(-/-), TRIF(-/-), and TLR4(-/-) DCs than in TLR2(-/-) DCs. Furthermore, C. jejuni induced interferon regulatory factor 3 phosphorylation and IFN-beta secretion by DCs in a TLR4-TRIF-dependent fashion, further demonstrating activation of this pathway by C. jejuni. Importantly, TLR2, TLR4, MyD88, and TRIF deficiencies all markedly impaired the Th1-priming ability of C. jejuni-infected DCs. Thus, our results show that cooperative signaling through the TLR4-MyD88 and TLR4-TRIF axes represents a novel mechanism mediating C. jejuni-induced inflammatory responses of DCs. To our knowledge, such a mechanism has not been demonstrated previously for an intact bacterium.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Campylobacter jejuni/inmunología , Células Dendríticas/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 4/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Animales , Citocinas/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Fosforilación , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/deficiencia
14.
BMC Microbiol ; 9: 57, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19296832

RESUMEN

BACKGROUND: Campylobacter jejuni infection produces a spectrum of clinical presentations in humans--including asymptomatic carriage, watery diarrhea, and bloody diarrhea--and has been epidemiologically associated with subsequent autoimmune neuropathies. This microorganism is genetically variable and possesses genetic mechanisms that may contribute to variability in nature. However, relationships between genetic variation in the pathogen and variation in disease manifestation in the host are not understood. We took a comparative experimental approach to explore differences among different C. jejuni strains and studied the effect of diet on disease manifestation in an interleukin-10 deficient mouse model. RESULTS: In the comparative study, C57BL/6 interleukin-10-/- mice were infected with seven genetically distinct C. jejuni strains. Four strains colonized the mice and caused disease; one colonized with no disease; two did not colonize. A DNA:DNA microarray comparison of the strain that colonized mice without disease to C. jejuni 11168 that caused disease revealed that putative virulence determinants, including loci encoding surface structures known to be involved in C. jejuni pathogenesis, differed from or were absent in the strain that did not cause disease. In the experimental study, the five colonizing strains were passaged four times in mice. For three strains, serial passage produced increased incidence and degree of pathology and decreased time to develop pathology; disease shifted from watery to bloody diarrhea. Mice kept on an ~6% fat diet or switched from an approximately 12% fat diet to an approximately 6% fat diet just before infection with a non-adapted strain also exhibited increased incidence and severity of disease and decreased time to develop disease, although the effects of diet were only statistically significant in one experiment. CONCLUSION: C. jejuni strain genetic background and adaptation of the strain to the host by serial passage contribute to differences in disease manifestations of C. jejuni infection in C57BL/6 IL-10-/- mice; differences in environmental factors such as diet may also affect disease manifestation. These results in mice reflect the spectrum of clinical presentations of C. jejuni gastroenteritis in humans and contribute to usefulness of the model in studying human disease.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/patogenicidad , Dieta , Enteritis/microbiología , Animales , Técnicas de Tipificación Bacteriana , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/patología , Campylobacter jejuni/clasificación , Campylobacter jejuni/genética , Análisis por Conglomerados , ADN Bacteriano/genética , Diarrea/etiología , Diarrea/microbiología , Modelos Animales de Enfermedad , Enteritis/inmunología , Enteritis/patología , Interleucina-10/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Longitud del Fragmento de Restricción , Pase Seriado , Virulencia
15.
Am J Vet Res ; 70(1): 49-56, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19119948

RESUMEN

OBJECTIVE: To evaluate dogs as a sentinel species for emergence of Lyme disease in a region undergoing invasion by Ixodes scapularis. SAMPLE POPULATION: 353 serum samples and 78 ticks obtained from dogs brought to 18 veterinary clinics located in the lower peninsula of Michigan from July 15, 2005, through August 15, 2005. PROCEDURES: Serum samples were evaluated for specific antibodies against Borrelia burgdorferi by use of 3 serologic assays. Ticks from dogs were subjected to PCR assays for detection of pathogens. RESULTS: Of 353 serum samples from dogs in 18 counties in 2005, only 2 (0.6%) contained western blot analysis-confirmed antibodies against B burgdorferi. Ten of 13 dogs with I scapularis were from clinics within or immediately adjacent to the known tick invasion zone. Six of 18 I scapularis and 12 of 60 noncompetent vector ticks were infected with B burgdorferi. No ticks were infected with Anaplasma phagocytophilum, and 3 were infected with Babesia spp. CONCLUSIONS AND CLINICAL RELEVANCE: Serosurvey in dogs was found to be ineffective in tracking early invasion dynamics of I scapularis in this area. Tick chemoprophylaxis likely reduces serosurvey sensitivity in dogs. Ticks infected with B burgdorferi were more common and widely dispersed than seropositive dogs. In areas of low tick density, use of dogs as a source of ticks is preferable to serosurvey for surveillance of emerging Lyme disease. IMPACT FOR HUMAN MEDICINE: By retaining ticks from dogs for identification and pathogen testing, veterinarians can play an important role in early detection in areas with increasing risk of Lyme disease.


Asunto(s)
Borrelia burgdorferi/crecimiento & desarrollo , Enfermedades de los Perros/epidemiología , Enfermedades de los Perros/parasitología , Ixodes/microbiología , Enfermedad de Lyme/epidemiología , Infestaciones por Garrapatas/veterinaria , Animales , Animales Domésticos/microbiología , Animales Domésticos/parasitología , Anticuerpos Antibacterianos/sangre , Western Blotting/veterinaria , Enfermedades de los Perros/microbiología , Perros , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/virología , Michigan/epidemiología , Vigilancia de Guardia/veterinaria , Estudios Seroepidemiológicos , Infestaciones por Garrapatas/epidemiología , Infestaciones por Garrapatas/microbiología
16.
J Neuroimmunol ; 337: 577048, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31678855

RESUMEN

The peripheral neuropathy Guillain-Barré Syndrome can follow Campylobacter jejuni infection when outer core lipooligosaccharides induce production of neurotoxic anti-ganglioside antibodies. We hypothesized that gut microbiota depletion with an antibiotic would increase C. jejuni colonization, severity of gastroenteritis, and GBS. Microbiota depletion increased C. jejuni colonization, invasion, and colitis with Type 1/17 T cells in gut lamina propria. It also stimulated Type 1/17 anti-C. jejuni and -antiganglioside-antibodies, Type 2 anti-C. jejuni and -antiganglioside antibodies, and neurologic phenotypes. Results indicate that both C. jejuni strain and gut microbiota affect development of inflammation and GBS and suggest that probiotics following C. jejuni infection may ameliorate inflammation and autoimmune disease.


Asunto(s)
Antibacterianos/toxicidad , Autoinmunidad/efectos de los fármacos , Infecciones por Campylobacter/patología , Colitis/patología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Autoinmunidad/fisiología , Infecciones por Campylobacter/inducido químicamente , Infecciones por Campylobacter/inmunología , Campylobacter jejuni/efectos de los fármacos , Campylobacter jejuni/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Femenino , Microbioma Gastrointestinal/fisiología , Síndrome de Guillain-Barré/inducido químicamente , Síndrome de Guillain-Barré/inmunología , Síndrome de Guillain-Barré/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/efectos de los fármacos , Microbiota/fisiología , Índice de Severidad de la Enfermedad
17.
Microbes Infect ; 10(12-13): 1316-24, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18725315

RESUMEN

Food-borne Campylobacter jejuni (Cj) is an important cause of enteritis. We showed that C57BL/6 and congenic interleukin (IL)-10(-/-) mice serve as models of Cj colonization and enteritis, respectively. Thus, C57BL/6 mice are resistant to Cj induced disease. Because dendritic cells (DCs) are central to regulating adaptive immune responses, we investigated the interaction of Cj with murine bone marrow-derived DCs (BM-DCs) to assess bacterial killing, DC activation, and the ability of Cj-infected BM-DCs to stimulate Campylobacter-specific T cell responses in vitro. BM-DCs challenged with Cj efficiently internalized and killed Cj 11168 and significantly upregulated surface MHC-II, CD40, CD80 and CD86 demonstrating a mature phenotype. Infected BM-DCs secreted significant amounts of tumor necrosis factor-alpha (TNF-alpha), IL-6 and IL-12p70. Formalin-killed Cj also induced maturation of BM-DCs with similar cytokine production but at a significantly lower magnitude than live bacteria. Maximal activation of murine BM-DCs required internalization of Cj; attachment alone was not sufficient to elicit significant responses. Also, various strains of Cj elicited different magnitudes of cytokine production from BM-DCs. Finally, in a coculture system, Cj-infected BM-DCs induced high level interferon-gamma (INF-gamma) production from CD4+T cells indicating Th1 polarization. Thus, DCs from resistant C57BL/6 mice initiate T cell responses against Cj.


Asunto(s)
Campylobacter jejuni/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células TH1/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Campylobacter jejuni/patogenicidad , Diferenciación Celular , Polaridad Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/citología , Interacciones Huésped-Patógeno , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL
18.
Environ Health Perspect ; 115(6): 856-64, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17589591

RESUMEN

BACKGROUND: A groundwater-associated outbreak affected approximately 1,450 residents and visitors of South Bass Island, Ohio, between July and September 2004. OBJECTIVES: To examine the microbiological quality of groundwater wells located on South Bass Island, we sampled 16 wells that provide potable water to public water systems 15-21 September 2004. METHODS: We tested groundwater wells for fecal indicators, enteric viruses and bacteria, and protozoa (Cryptosporidium and Giardia). The hydrodynamics of Lake Erie were examined to explore the possible surface water-groundwater interactions. RESULTS: All wells were positive for both total coliform and Escherichia coli. Seven wells tested positive for enterococci and Arcobacter (an emerging bacterial pathogen), and F(+)-specific coliphage was present in four wells. Three wells were positive for all three bacterial indicators, coliphages, and Arcobacter; adenovirus DNA was recovered from two of these wells. We found a cluster of the most contaminated wells at the southeast side of the island. CONCLUSIONS: Massive groundwater contamination on the island was likely caused by transport of microbiological contaminants from wastewater treatment facilities and septic tanks to the lake and the subsurface, after extreme precipitation events in May-July 2004. This likely raised the water table, saturated the subsurface, and along with very strong Lake Erie currents on 24 July, forced a surge in water levels and rapid surface water-groundwater interchange throughout the island. Landsat images showed massive influx of organic material and turbidity surrounding the island before the peak of the outbreak. These combinations of factors and information can be used to examine vulnerabilities in other coastal systems. Both wastewater and drinking water issues are now being addressed by the Ohio Environmental Protection Agency and the Ohio Department of Health.


Asunto(s)
Bacterias/aislamiento & purificación , Monitoreo del Ambiente/estadística & datos numéricos , Virus/aislamiento & purificación , Microbiología del Agua , Abastecimiento de Agua , Animales , Cryptosporidium/aislamiento & purificación , Cartilla de ADN/genética , Geografía , Giardia/aislamiento & purificación , Microscopía Fluorescente , Ohio , Polimorfismo de Longitud del Fragmento de Restricción , Virus/genética , Movimientos del Agua
19.
Vet Parasitol ; 149(1-2): 43-55, 2007 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-17706872

RESUMEN

Sarcocystis neurona is an important protozoal pathogen because it causes the serious neurological disease equine protozoal myeloencephalitis (EPM). The capacity of this organism to cause a wide spectrum of neurological signs in horses and the broad geographic distribution of observed cases in the Americas drive the need for sensitive, reliable and rapid typing methods to characterize strains. Various molecular methods have been developed and used to diagnose EPM due to S. neurona, to identify S. neurona isolates and to determine the heterogeneity and evolutionary relatedness within this species and related Sarcocystis spp. These methods included sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), immuno-fluorescent assay (IFA), slide agglutination test (SAT), SnSAG-specific ELISA, random amplified polymorphic DNA (RAPD), PCR-based restriction fragment length polymorphism (RFLP), amplified fragment length polymorphism (AFLP) fingerprinting, and sequence analysis of surface protein genes, ribosomal genes, microsatellite alleles and other molecular markers. Here, the utility of these molecular methods is reviewed and evaluated with respect to the need for molecular approaches that utilize well-characterized polymorphic, simple, independent, and stable genetic markers. These tools have the potential to add to knowledge of the genetic population structure of S. neurona and to provide new insights into the pathogenesis of EPM and S. neurona epidemiology. In particular, these methods provide new tools to address the hypothesis that particular genetic variants are associated with adverse clinical outcomes (severe pathotypes). The ultimate goal is to utilize them in future studies to improve treatment and prevention strategies.


Asunto(s)
Encefalomielitis/veterinaria , Enfermedades de los Caballos/diagnóstico , Sarcocystis/clasificación , Sarcocystis/genética , Sarcocistosis/veterinaria , Animales , Encefalomielitis/diagnóstico , Encefalomielitis/epidemiología , Enfermedades de los Caballos/epidemiología , Caballos , Técnicas de Diagnóstico Molecular/tendencias , Técnicas de Diagnóstico Molecular/veterinaria , Sarcocistosis/diagnóstico , Sarcocistosis/epidemiología
20.
Anim Health Res Rev ; 18(2): 99-111, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29665882

RESUMEN

Campylobacter jejuni is an important zoonotic pathogen recently designated a serious antimicrobial resistant (AR) threat. While most patients with C. jejuni experience hemorrhagic colitis, serious autoimmune conditions can follow including inflammatory bowel disease (IBD) and the acute neuropathy Guillain Barré Syndrome (GBS). This review examines inter-relationships among factors mediating C. jejuni diarrheal versus autoimmune disease especially AR C. jejuni and microbiome shifts. Because both susceptible and AR C. jejuni are acquired from animals or their products, we consider their role in harboring strains. Inter-relationships among factors mediating C. jejuni colonization, diarrheal and autoimmune disease include C. jejuni virulence factors and AR, the enteric microbiome, and host responses. Because AR C. jejuni have been suggested to affect the severity of disease, length of infections and propensity to develop GBS, it is important to understand how these interactions occur when strains are under selection by antimicrobials. More work is needed to elucidate host-pathogen interactions of AR C. jejuni compared with susceptible strains and how AR C. jejuni are maintained and evolve in animal reservoirs and the extent of transmission to humans. These knowledge gaps impair the development of effective strategies to prevent the emergence of AR C. jejuni in reservoir species and human populations.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/efectos de los fármacos , Farmacorresistencia Microbiana , Microbiota , Animales , Síndrome de Guillain-Barré , Interacciones Huésped-Patógeno , Humanos , Factores de Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA