Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(2): e2316540120, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38170751

RESUMO

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using 6-wk-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni, ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within 2 to 3 d of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP, which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter (lctP) led to identification of a putative thiol-based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides better insights into the pathogenicity of C. jejuni.


Assuntos
Infecções por Campylobacter , Campylobacter jejuni , Animais , Humanos , Camundongos , Ácido Láctico/metabolismo , Campylobacter jejuni/genética , Furões , Transportadores de Ácidos Monocarboxílicos
2.
bioRxiv ; 2023 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-37873437

RESUMO

How the microaerobic pathogen Campylobacter jejuni establishes its niche and expands in the gut lumen during infection is poorly understood. Using six-week-old ferrets as a natural disease model, we examined this aspect of C. jejuni pathogenicity. Unlike mice, which require significant genetic or physiological manipulation to become colonized with C. jejuni , ferrets are readily infected without the need to disarm the immune system or alter the gut microbiota. Disease after C. jejuni infection in ferrets reflects closely how human C. jejuni infection proceeds. Rapid growth of C. jejuni and associated intestinal inflammation was observed within two-three days of infection. We observed pathophysiological changes that were noted by cryptic hyperplasia through the induction of tissue repair systems, accumulation of undifferentiated amplifying cells on the colon surface, and instability of HIF-1α in colonocytes, which indicated increased epithelial oxygenation. Metabolomic analysis demonstrated that lactate levels in colon content were elevated in infected animals. A C. jejuni mutant lacking lctP , which encodes an L-lactate transporter, was significantly decreased for colonization during infection. Lactate also influences adhesion and invasion by C. jejuni to a colon carcinoma cell line (HCT116). The oxygenation required for expression of lactate transporter ( lctP ) led to discovery of a putative thiol based redox switch regulator (LctR) that may repress lctP transcription under anaerobic conditions. Our work provides new insights into the pathogenicity of C. jejuni . Significance: There is a gap in knowledge about the mechanisms by which C. jejuni populations expand during infection. Using an animal model which accurately reflects human infection without the need to alter the host microbiome or the immune system prior to infection, we explored pathophysiological alterations of the gut after C. jejuni infection. Our study identified the gut metabolite L-lactate as playing an important role as a growth substrate for C. jejuni during acute infection. We identified a DNA binding protein, LctR, that binds to the lctP promoter and may repress lctP expression, resulting in decreased lactate transport under low oxygen levels. This work provides new insights about C. jejuni pathogenicity.

3.
J Bacteriol ; 202(7)2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-31932316

RESUMO

Campylobacter jejuni causes acute gastroenteritis worldwide and is transmitted primarily through poultry, in which it is often a commensal member of the intestinal microbiota. Previous transcriptome sequencing (RNA-Seq) experiment showed that transcripts from an operon encoding a high-affinity phosphate transporter (PstSCAB) of C. jejuni were among the most abundant when the bacterium was grown in chickens. Elevated levels of the pstSCAB mRNA were also identified in an RNA-Seq experiment from human infection studies. In this study, we explore the role of PstSCAB in the biology and colonization potential of C. jejuni Our results demonstrate that cells lacking PstSCAB survive poorly in stationary phase, in nutrient-limiting media, and under osmotic conditions reflective of those in the chicken. Polyphosphate levels in the mutant cells were elevated at stationary phase, consistent with alterations in expression of polyphosphate metabolism genes. The mutant strain was highly attenuated for colonization of newly hatched chicks, with levels of bacteria at several orders of magnitude below wild-type levels. Mutant and wild type grew similarly in complex media, but the pstS::kan mutant exhibited a significant growth defect in minimal medium supplemented with l-lactate, postulated as a carbon source in vivo Poor growth in lactate correlated with diminished expression of acetogenesis pathway genes previously demonstrated as important for colonizing chickens. The phosphate transport system is thus essential for diverse aspects of C. jejuni physiology and in vivo fitness and survival.IMPORTANCECampylobacter jejuni causes millions of human gastrointestinal infections annually, with poultry a major source of infection. Due to the emergence of multidrug resistance in C. jejuni, there is need to identify alternative ways to control this pathogen. Genes encoding the high-affinity phosphate transporter PstSCAB are highly expressed by C. jejuni in chickens and humans. In this study, we address the role of PstSCAB on chicken colonization and other C. jejuni phenotypes. PstSCAB is required for colonization in chicken, metabolism and survival under different stress responses, and during growth on lactate, a potential growth substrate in chickens. Our study highlights that PstSCAB may be an effective target to develop mechanisms for controlling bacterial burden in both chicken and human.


Assuntos
Infecções por Campylobacter/veterinária , Campylobacter jejuni/fisiologia , Galinhas/microbiologia , Ácido Láctico/metabolismo , Proteínas de Transporte de Fosfato/genética , Doenças das Aves Domésticas/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Metabolômica/métodos , Mutação , Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/metabolismo , Estresse Fisiológico
5.
J Biol Chem ; 289(12): 8007-18, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24394413

RESUMO

Despite the importance of Campylobacter jejuni as a pathogen, little is known about the fundamental aspects of its peptidoglycan (PG) structure and factors modulating its helical morphology. A PG dl-carboxypeptidase Pgp1 essential for maintenance of C. jejuni helical shape was recently identified. Bioinformatic analysis revealed the CJJ81176_0915 gene product as co-occurring with Pgp1 in several organisms. Deletion of cjj81176_0915 (renamed pgp2) resulted in straight morphology, representing the second C. jejuni gene affecting cell shape. The PG structure of a Δpgp2 mutant showed an increase in tetrapeptide-containing muropeptides and a complete absence of tripeptides, consistent with ld-carboxypeptidase activity, which was confirmed biochemically. PG analysis of a Δpgp1Δpgp2 double mutant demonstrated that Pgp2 activity is required to generate the tripeptide substrate for Pgp1. Loss of pgp2 affected several pathogenic properties; the deletion strain was defective for motility in semisolid agar, biofilm formation, and fluorescence on calcofluor white. Δpgp2 PG also caused decreased stimulation of the human nucleotide-binding oligomerization domain 1 (Nod1) proinflammatory mediator in comparison with wild type, as expected from the reduction in muropeptide tripeptides (the primary Nod1 agonist) in the mutant; however, these changes did not alter the ability of the Δpgp2 mutant strain to survive within human epithelial cells or to elicit secretion of IL-8 from epithelial cells after infection. The pgp2 mutant also showed significantly reduced fitness in a chick colonization model. Collectively, these analyses enhance our understanding of C. jejuni PG maturation and help to clarify how PG structure and cell shape impact pathogenic attributes.


Assuntos
Infecções por Campylobacter/microbiologia , Campylobacter jejuni/citologia , Campylobacter jejuni/enzimologia , Carboxipeptidases/metabolismo , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Biofilmes/crescimento & desenvolvimento , Campylobacter jejuni/patogenicidade , Campylobacter jejuni/fisiologia , Carboxipeptidases/genética , Linhagem Celular , Deleção de Genes , Humanos , Peptidoglicano/química , Peptidoglicano/metabolismo
6.
PLoS Pathog ; 8(3): e1002602, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22457624

RESUMO

The impact of bacterial morphology on virulence and transmission attributes of pathogens is poorly understood. The prevalent enteric pathogen Campylobacter jejuni displays a helical shape postulated as important for colonization and host interactions. However, this had not previously been demonstrated experimentally. C. jejuni is thus a good organism for exploring the role of factors modulating helical morphology on pathogenesis. We identified an uncharacterized gene, designated pgp1 (peptidoglycan peptidase 1), in a calcofluor white-based screen to explore cell envelope properties important for C. jejuni virulence and stress survival. Bioinformatics showed that Pgp1 is conserved primarily in curved and helical bacteria. Deletion of pgp1 resulted in a striking, rod-shaped morphology, making pgp1 the first C. jejuni gene shown to be involved in maintenance of C. jejuni cell shape. Pgp1 contributes to key pathogenic and cell envelope phenotypes. In comparison to wild type, the rod-shaped pgp1 mutant was deficient in chick colonization by over three orders of magnitude and elicited enhanced secretion of the chemokine IL-8 in epithelial cell infections. Both the pgp1 mutant and a pgp1 overexpressing strain - which similarly produced straight or kinked cells - exhibited biofilm and motility defects. Detailed peptidoglycan analyses via HPLC and mass spectrometry, as well as Pgp1 enzyme assays, confirmed Pgp1 as a novel peptidoglycan DL-carboxypeptidase cleaving monomeric tripeptides to dipeptides. Peptidoglycan from the pgp1 mutant activated the host cell receptor Nod1 to a greater extent than did that of wild type. This work provides the first link between a C. jejuni gene and morphology, peptidoglycan biosynthesis, and key host- and transmission-related characteristics.


Assuntos
Campylobacter jejuni/enzimologia , Campylobacter jejuni/genética , Genes Bacterianos , Interações Hospedeiro-Patógeno/genética , Peptídeo Hidrolases/metabolismo , Peptidoglicano/biossíntese , Animais , Campylobacter jejuni/patogenicidade , Linhagem Celular , Forma Celular/fisiologia , Galinhas , Cromatografia Líquida de Alta Pressão , Células Epiteliais/microbiologia , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos
7.
J Bacteriol ; 194(9): 2342-54, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22343300

RESUMO

Campylobacter jejuni commensally colonizes the cecum of birds. The RacR (reduced ability to colonize) response regulator was previously shown to be important in avian colonization. To explore the means by which RacR and its cognate sensor kinase RacS may modulate C. jejuni physiology and colonization, ΔracR and ΔracS mutations were constructed in the invasive, virulent strain 81-176, and extensive phenotypic analyses were undertaken. Both the ΔracR and ΔracS mutants exhibited a ~100-fold defect in chick colonization despite no (ΔracS) or minimal (ΔracR) growth defects at 42 °C, the avian body temperature. Each mutant was defective for colony formation at 44°C and in the presence of 0.8% NaCl, both of which are stresses associated with the heat shock response. Promoter-reporter and real-time quantitative PCR (RT-qPCR) analyses revealed that RacR activates racRS and represses dnaJ. Although disregulation of several other heat shock genes was not observed at 38°C, the ΔracR and ΔracS mutants exhibited diminished upregulation of these genes upon a rapid temperature upshift. Furthermore, the ΔracR and ΔracS mutants displayed increased length heterogeneity during exponential growth, with a high proportion of filamented bacteria. Filamented bacteria had reduced swimming speed and were defective for invasion of Caco-2 epithelial cells. Soft-agar studies also revealed that the loss of racR or racS resulted in whole-population motility defects in viscous medium. These findings reveal new roles for RacRS in C. jejuni physiology, each of which is likely important during colonization of the avian host.


Assuntos
Proteínas de Bactérias/metabolismo , Campylobacter jejuni/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Resposta ao Choque Térmico/fisiologia , Ágar , Animais , Proteínas de Bactérias/genética , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/veterinária , Campylobacter jejuni/citologia , Campylobacter jejuni/genética , Galinhas , Meios de Cultura , Células Epiteliais/microbiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Intestinos/citologia , Intestinos/microbiologia , Movimento , Mutação
8.
J Bacteriol ; 191(1): 375-87, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18952801

RESUMO

HAMP domains, found in many bacterial signal transduction proteins, generally transmit an intramolecular signal between an extracellular sensory domain and an intracellular signaling domain. Studies of HAMP domains in proteins where both the input and output signals occur intracellularly are limited to those of the Aer energy taxis receptor of Escherichia coli, which has both a HAMP domain and a sensory PAS domain. Campylobacter jejuni has an energy taxis system consisting of the domains of Aer divided between two proteins, CetA (HAMP domain containing) and CetB (PAS domain containing). In this study, we found that the CetA HAMP domain differs significantly from that of Aer in the predicted secondary structure. Using similarity searches, we identified 55 pairs of HAMP/PAS proteins encoded by adjacent genes in a diverse group of microorganisms. We propose that these HAMP/PAS pairs form a new family of bipartite energy taxis receptors. Within these proteins, we identified nine residues in the HAMP domain and proximal signaling domain that are highly conserved, at least three of which are required for CetA function. Additionally, we demonstrated that CetA contributes to the invasion of human epithelial cells by C. jejuni, while CetB does not. This finding supports the hypothesis that members of HAMP/PAS pairs possess the capacity to act independently of each other in cellular traits other than energy taxis.


Assuntos
Campylobacter jejuni/genética , Proteínas de Transporte/genética , Sequência de Aminoácidos , Proteínas Arqueais/química , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Archaeoglobales/genética , Campylobacter jejuni/metabolismo , Campylobacter jejuni/patogenicidade , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Quimiotaxia/genética , Sequência Conservada , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Transdução de Sinais/genética
9.
Infect Immun ; 74(12): 6957-64, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17000721

RESUMO

Intestinal M cells bear a receptor for secretory immunoglobulin A (IgA) (sIgA) facing the lumen of the epithelial surfaces. Cells bearing this receptor are also found throughout an experimental monolayer consisting of polarized Caco-2 cells, a colon adenocarcinoma cell line. The presence of antibodies (mainly sIgA) in the lumen of the small intestine led us to explore the participation of the sIgA receptor and antibodies in the interaction of Caco-2-associated M-like cells with the mucosal pathogen Vibrio cholerae. Here, we demonstrate that sIgA antibodies isolated from pooled healthy human colostrums, as well as IgG from pooled healthy human serum, can recognize V. cholerae. Furthermore, opsonization enhances M-like-cell transcytosis of V. cholerae strains. We also show that the cholera toxin (CT) receptor ganglioside GM(1) colocalizes with the sIgA receptor in cells of the epithelial monolayer. Both sIgA and IgG antibodies compete for the attachment of soluble CT subunit B to immobilized GM(1). Our results indicate that in this in vitro model system of intestinal epithelia, human sIgA and IgG contribute to the uptake of V. cholerae by M-like cells, probably through an interaction with GM(1). Our results support previous findings of others showing that sIgA can act as an endogenous adjuvant and that sIgA is important for the antigen-sampling function of M cells.


Assuntos
Endocitose/imunologia , Gangliosídeo G(M1)/metabolismo , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Receptores de Superfície Celular/metabolismo , Vibrio cholerae/imunologia , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/isolamento & purificação , Anticorpos Antibacterianos/farmacologia , Células Cultivadas , Toxina da Cólera/genética , Colostro/imunologia , Endocitose/efeitos dos fármacos , Gangliosídeo G(M1)/análise , Humanos , Imunidade nas Mucosas , Imunoglobulina A Secretora/isolamento & purificação , Imunoglobulina A Secretora/farmacologia , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Imunoglobulina G/farmacologia , Mucosa Intestinal/química , Intestino Delgado/imunologia , Receptores de Superfície Celular/análise , Receptores Fc/análise , Receptores Fc/metabolismo , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/genética
10.
Infect Immun ; 74(8): 4715-23, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16861659

RESUMO

Campylobacter jejuni has an N-linked protein glycosylation pathway that is required for efficient cell invasion and chick gastrointestinal colonization by the microbe. In this study, we constructed insertion mutants of 22 putative glycoprotein genes and examined the ability of each to invade the human intestinal epithelial cell line INT-407. Among the mutants tested, one carrying an insertion in Cj1496c was defective for invasion into INT-407 cells; this defect was also observed in an in-frame deletion mutant of Cj1496c (delta Cj1496c). The delta Cj1496c mutant C. jejuni also showed a reduced ability to colonize chick ceca. Site-specific mutagenesis combined with Western blot analysis suggested that the Cj1496c protein is glycosylated at N73 and N169. However, the delta Cj1496c mutant expressing a nonglycosylated form of Cj1496c exhibited levels of invasion and colonization equivalent to those of the parent strain, suggesting that glycans are not directly involved in the function of Cj1496c.


Assuntos
Campylobacter jejuni/patogenicidade , Galinhas/microbiologia , Células Epiteliais/microbiologia , Trato Gastrointestinal/microbiologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Animais , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/genética , Campylobacter jejuni/crescimento & desenvolvimento , Ceco/microbiologia , Linhagem Celular , Deleção de Genes , Glicosilação , Humanos , Intestinos/citologia , Intestinos/microbiologia , Mutagênese Sítio-Dirigida
11.
Infect Immun ; 72(2): 623-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14742501

RESUMO

Streptococcus pyogenes with null mutations in the csrRS regulatory locus are highly virulent in mice due to derepression of hyaluronic acid capsule synthesis and exotoxins, e.g., streptolysin S (SLS) and pyrogenic exotoxin B (SpeB). We generated derivatives of a DeltacsrRS strain that also carry deletions in hasAB (leading to an acapsular phenotype) or in sagA (phenotypically SLS-) or an interruption of speB (SpeB-) to test the relative contributions of these factors to the development of necrotic skin lesions. Inoculation of 2 x 10(6) to 4 x 10(6) CFU of either acapsular or SLS- strains into hairless mice resulted in lesions approximately 70% smaller than those of the DeltacsrRS parent strain. Elimination of SLS also reduced lethality from 100% to 0% at this inoculum (P < 10(-7); Fisher exact test). In contrast, SLS+ SpeB- mutants yielded lesions that were only 41% smaller than the parent strain (t = 2.2; P = 0.04), but only 3 the 17 lesions had dermal sloughing (P = 10(-5)). The nonulcerative lesions associated with SpeB- strains appeared pale with surrounding erythema. We conclude that capsule and SLS contribute to the subcutaneous spread of S. pyogenes and to a fatal outcome of infection. SpeB facilitates early dermal ulceration but has minor influence on lesion size and mortality. Large ulcerative lesions are observed only when both toxins are present.


Assuntos
Proteínas de Bactérias/fisiologia , Cisteína Endopeptidases/fisiologia , Proteínas Repressoras/fisiologia , Dermatopatias Infecciosas/etiologia , Infecções Estreptocócicas/etiologia , Streptococcus pyogenes/patogenicidade , Estreptolisinas/fisiologia , Fatores de Virulência/fisiologia , Animais , Ácido Hialurônico/biossíntese , Masculino , Camundongos , Camundongos Pelados
12.
Mem. Inst. Oswaldo Cruz ; 93(5): 567-76, Sept.-Oct. 1998. graf
Artigo em Inglês | LILACS | ID: lil-217853

RESUMO

Evolutionary theory may contribute to practical solutions for control of disease by identifying interventions that may cause pathogens to evolve to reduce virulence. Theory predicts, for example, that pathogens transmitted by water or arthropod vectors should evolve to relatively high levels of virulence because such pathogens can gain the evolutionary benefits of relatively high levels of host exploitation while paying little price from host illness. The entrance of Vibrio cholerae into South America in 1991 has generated a natural experiment that allows testing of this idea by determining whether geographic and temporal variations in toxigenicity correspond to variation in the potencial for waterborne transmission. Preliminary studies show such correspondences: toxigenicity is negatively associated with access to uncontaminated water in Brazil; and in Chile, where the potential for waterborne transmission is particularly low, toxigenicity of strains declined between 1991 and 1998. In theory vector-proofing of houses should be similarly associated with benignity of vectorborne pathogens, such as the agents of dengue, malaria, and Chagas'disease. These preliminary studies draw attention to the need for definitive prospective experiments to determine whether interventions such as provisioning of uncontaminated water and vector -proofing of houses cause evolutionary reductions in virulence.


Assuntos
Controle de Doenças Transmissíveis , Doenças Transmissíveis/microbiologia , Virulência/imunologia , Água/parasitologia , Brasil , Transmissão de Doença Infecciosa , Vetores de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA