Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 578(7796): 582-587, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051588

RESUMEN

Addressing the ongoing antibiotic crisis requires the discovery of compounds with novel mechanisms of action that are capable of treating drug-resistant infections1. Many antibiotics are sourced from specialized metabolites produced by bacteria, particularly those of the Actinomycetes family2. Although actinomycete extracts have traditionally been screened using activity-based platforms, this approach has become unfavourable owing to the frequent rediscovery of known compounds. Genome sequencing of actinomycetes reveals an untapped reservoir of biosynthetic gene clusters, but prioritization is required to predict which gene clusters may yield promising new chemical matter2. Here we make use of the phylogeny of biosynthetic genes along with the lack of known resistance determinants to predict divergent members of the glycopeptide family of antibiotics that are likely to possess new biological activities. Using these predictions, we uncovered two members of a new functional class of glycopeptide antibiotics-the known glycopeptide antibiotic complestatin and a newly discovered compound we call corbomycin-that have a novel mode of action. We show that by binding to peptidoglycan, complestatin and corbomycin block the action of autolysins-essential peptidoglycan hydrolases that are required for remodelling of the cell wall during growth. Corbomycin and complestatin have low levels of resistance development and are effective in reducing bacterial burden in a mouse model of skin MRSA infection.


Asunto(s)
Antibacterianos , Descubrimiento de Drogas , Péptidos Cíclicos , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Actinobacteria/química , Actinobacteria/genética , Actinobacteria/metabolismo , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Vías Biosintéticas/genética , Pared Celular/metabolismo , Clorofenoles/química , Clorofenoles/metabolismo , Clorofenoles/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Familia de Multigenes , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacología , Filogenia , Piel/microbiología , Infecciones Estafilocócicas/microbiología
2.
Nat Chem Biol ; 19(11): 1342-1350, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37231267

RESUMEN

Acinetobacter baumannii is a nosocomial Gram-negative pathogen that often displays multidrug resistance. Discovering new antibiotics against A. baumannii has proven challenging through conventional screening approaches. Fortunately, machine learning methods allow for the rapid exploration of chemical space, increasing the probability of discovering new antibacterial molecules. Here we screened ~7,500 molecules for those that inhibited the growth of A. baumannii in vitro. We trained a neural network with this growth inhibition dataset and performed in silico predictions for structurally new molecules with activity against A. baumannii. Through this approach, we discovered abaucin, an antibacterial compound with narrow-spectrum activity against A. baumannii. Further investigations revealed that abaucin perturbs lipoprotein trafficking through a mechanism involving LolE. Moreover, abaucin could control an A. baumannii infection in a mouse wound model. This work highlights the utility of machine learning in antibiotic discovery and describes a promising lead with targeted activity against a challenging Gram-negative pathogen.


Asunto(s)
Acinetobacter baumannii , Aprendizaje Profundo , Animales , Ratones , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple , Pruebas de Sensibilidad Microbiana
3.
Infect Immun ; 91(1): e0050522, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36511702

RESUMEN

The NleGs are the largest family of type 3 secreted effectors in attaching and effacing (A/E) pathogens, such as enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli, and Citrobacter rodentium. NleG effectors contain a conserved C-terminal U-box domain acting as a ubiquitin protein ligase and target host proteins via a variable N-terminal portion. The specific roles of these effectors during infection remain uncertain. Here, we demonstrate that the three NleG effectors-NleG1Cr, NleG7Cr, and NleG8Cr-encoded by C. rodentium DBS100 play distinct roles during infection in mice. Using individual nleGCr knockout strains, we show that NleG7Cr contributes to bacterial survival during enteric infection while NleG1Cr promotes the expression of diarrheal symptoms and NleG8Cr contributes to accelerated lethality in susceptible mice. Furthermore, the NleG8Cr effector contains a C-terminal PDZ domain binding motif that enables interaction with the host protein GOPC. Both the PDZ domain binding motif and the ability to engage with host ubiquitination machinery via the intact U-box domain proved to be necessary for NleG8Cr function, contributing to the observed phenotype during infection. We also establish that the PTZ binding motif in the EHEC NleG8 (NleG8Ec) effector, which shares 60% identity with NleG8Cr, is engaged in interactions with human GOPC. The crystal structure of the NleG8Ec C-terminal peptide in complex with the GOPC PDZ domain, determined to 1.85 Å, revealed a conserved interaction mode similar to that observed between GOPC and eukaryotic PDZ domain binding motifs. Despite these common features, nleG8Ec does not complement the ΔnleG8Cr phenotype during infection, revealing functional diversification between these NleG effectors.


Asunto(s)
Infecciones por Enterobacteriaceae , Escherichia coli Enterohemorrágica , Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Humanos , Animales , Ratones , Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/microbiología , Transporte Biológico , Proteínas de Escherichia coli/genética , Escherichia coli Enteropatógena/genética , Escherichia coli Enterohemorrágica/genética , Proteínas de la Matriz de Golgi/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
4.
PLoS Pathog ; 17(5): e1009532, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33984072

RESUMEN

Bacteria inhabit diverse environmental niches and consequently must modulate their metabolism to adapt to stress. The nucleotide second messengers guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp) (collectively referred to as (p)ppGpp) are essential for survival during nutrient starvation. (p)ppGpp is synthesized by the RelA-SpoT homologue (RSH) protein family and coordinates the control of cellular metabolism through its combined effect on over 50 proteins. While the role of (p)ppGpp has largely been associated with nutrient limitation, recent studies have shown that (p)ppGpp and related nucleotides have a previously underappreciated effect on different aspects of bacterial physiology, such as maintaining cellular homeostasis and regulating bacterial interactions with a host, other bacteria, or phages. (p)ppGpp produced by pathogenic bacteria facilitates the evasion of host defenses such as reactive nitrogen intermediates, acidic pH, and the complement system. Additionally, (p)ppGpp and pyrophosphorylated derivatives of canonical adenosine nucleotides called (p)ppApp are emerging as effectors of bacterial toxin proteins. Here, we review the RSH protein family with a focus on its unconventional roles during host infection and bacterial competition.


Asunto(s)
Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Fenómenos Fisiológicos Bacterianos , Proteínas Bacterianas/metabolismo , Difosfatos/metabolismo , Nucleótidos/metabolismo , Estrés Fisiológico , Animales , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Fosforilación
5.
Infect Immun ; 89(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33139383

RESUMEN

The stringent response is an essential mechanism of metabolic reprogramming during environmental stress that is mediated by the nucleotide alarmones guanosine tetraphosphate and pentaphosphate [(p)ppGpp]. In addition to physiological adaptations, (p)ppGpp also regulates virulence programs in pathogenic bacteria, including Salmonella enterica serovar Typhimurium. S Typhimurium is a common cause of acute gastroenteritis, but it may also spread to systemic tissues, resulting in severe clinical outcomes. During infection, S Typhimurium encounters a broad repertoire of immune defenses that it must evade for successful host infection. Here, we examined the role of the stringent response in S Typhimurium resistance to complement-mediated killing and found that the (p)ppGpp synthetase-hydrolase, SpoT, is required for bacterial survival in human serum. We identified the nucleotide hydrolase, PpnN, as a target of the stringent response that is required to promote bacterial fitness in serum. Using chromatography and mass spectrometry, we show that PpnN hydrolyzes purine and pyrimidine monophosphates to generate free nucleobases and ribose 5'-phosphate, and that this metabolic activity is required for conferring resistance to complement killing. In addition to PpnN, we show that (p)ppGpp is required for the biosynthesis of the very long and long O-antigen in the outer membrane, known to be important for complement resistance. Our results provide new insights into the role of the stringent response in mediating evasion of the innate immune system by pathogenic bacteria.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Ligasas/inmunología , N-Glicosil Hidrolasas/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Virulencia/genética , Virulencia/inmunología , Regulación Bacteriana de la Expresión Génica , Variación Genética , Humanos , Inmunidad Innata , Ligasas/genética , N-Glicosil Hidrolasas/genética , Serogrupo
6.
Am J Physiol Endocrinol Metab ; 321(3): E338-E350, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34280051

RESUMEN

Obesity is associated with metabolic, immunological, and infectious disease comorbidities, including an increased risk of enteric infection and inflammatory bowel disease such as Crohn's disease (CD). Expansion of intestinal pathobionts such as adherent-invasive Escherichia coli (AIEC) is a common dysbiotic feature of CD, which is amplified by prior use of oral antibiotics. Although high-fat, high-sugar diets are associated with dysbiotic expansion of E. coli, it is unknown if the content of fat or another dietary component in obesogenic diets is sufficient to promote AIEC expansion. Here, we found that administration of an antibiotic combined with feeding mice an obesogenic low-fiber, high-sucrose, high-fat diet (HFD) that is typically used in rodent-obesity studies promoted AIEC intestinal expansion. Even a short-term (i.e., 1 day) pulse of HFD feeding before infection was sufficient to promote AIEC expansion, indicating that the magnitude of obesity was not the main driver of AIEC expansion. Controlled-diet experiments demonstrated that neither dietary fat nor sugar were the key determinants of AIEC colonization, but that lowering dietary fiber from approximately 13% to 5%-6% was sufficient to promote the intestinal expansion of AIEC when combined with antibiotics in mice. When combined with antibiotics, lowering fiber promoted AIEC intestinal expansion to a similar extent as widely used HFDs in mice. However, lowering dietary fiber was sufficient to promote AIEC intestinal expansion without affecting body mass. Our results show that low dietary fiber combined with oral antibiotics are environmental factors that promote the expansion of Crohn's disease-associated pathobionts in the gut.NEW & NOTEWORTHY It is commonly thought that obesity or a high-fat diet alters pathogenic bacteria and promotes inflammatory gut diseases. We found that lower dietary fiber is a key factor that expands a gut pathobiont linked to Crohn's disease, independent of obesity status in mice.


Asunto(s)
Enfermedad de Crohn/microbiología , Fibras de la Dieta/administración & dosificación , Intestinos/microbiología , Obesidad/microbiología , Animales , Escherichia coli/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Proc Natl Acad Sci U S A ; 115(40): 10004-10009, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30217892

RESUMEN

The pathogenic strategy of Escherichia coli and many other gram-negative pathogens relies on the translocation of a specific set of proteins, called effectors, into the eukaryotic host cell during infection. These effectors act in concert to modulate host cell processes in favor of the invading pathogen. Injected by the type III secretion system (T3SS), the effector arsenal of enterohemorrhagic E. coli (EHEC) O157:H7 features at least eight individual NleG effectors, which are also found across diverse attaching and effacing pathogens. NleG effectors share a conserved C-terminal U-box E3 ubiquitin ligase domain that engages with host ubiquitination machinery. However, their specific functions and ubiquitination targets have remained uncharacterized. Here, we identify host proteins targeted for ubiquitination-mediated degradation by two EHEC NleG family members, NleG5-1 and NleG2-3. NleG5-1 localizes to the host cell nucleus and targets the MED15 subunit of the Mediator complex, while NleG2-3 resides in the host cytosol and triggers degradation of Hexokinase-2 and SNAP29. Our structural studies of NleG5-1 reveal a distinct N-terminal α/ß domain that is responsible for interacting with host protein targets. The core of this domain is conserved across the NleG family, suggesting this domain is present in functionally distinct NleG effectors, which evolved diversified surface residues to interact with specific host proteins. This is a demonstration of the functional diversification and the range of host proteins targeted by the most expanded effector family in the pathogenic arsenal of E. coli.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Escherichia coli O157 , Proteínas de Escherichia coli , Infecciones por Escherichia coli/patología , Escherichia coli O157/química , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Células HeLa , Hexoquinasa/metabolismo , Humanos , Complejo Mediador/metabolismo , Dominios Proteicos , Proteolisis , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Células U937
8.
PLoS Pathog ; 14(8): e1007224, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30118511

RESUMEN

Enteropathogenic Escherichia coli (EPEC) use a needle-like injection apparatus known as the type III secretion system (T3SS) to deliver protein effectors into host cells. Effector translocation is highly stratified in EPEC with the translocated intimin receptor (Tir) being the first effector delivered into the host. CesT is a multi-cargo chaperone that is required for the secretion of Tir and at least 9 other effectors. However, the structural and mechanistic basis for differential effector recognition by CesT remains unclear. Here, we delineated the minimal CesT-binding region on Tir to residues 35-77 and determined the 2.74 Å structure of CesT bound to an N-terminal fragment of Tir. Our structure revealed that the CesT-binding region in the N-terminus of Tir contains an additional conserved sequence, distinct from the known chaperone-binding ß-motif, that we termed the CesT-extension motif because it extends the ß-sheet core of CesT. This motif is also present in the C-terminus of Tir that we confirmed to be a unique second CesT-binding region. Point mutations that disrupt CesT-binding to the N- or C-terminus of Tir revealed that the newly identified carboxy-terminal CesT-binding region was required for efficient Tir translocation into HeLa cells and pedestal formation. Furthermore, the CesT-extension motif was identified in the N-terminal region of NleH1, NleH2, and EspZ, and mutations that disrupt this motif reduced translocation of these effectors, and in some cases, overall effector stability, thus validating the universality of this CesT-extension motif. The presence of two CesT-binding regions in Tir, along with the presence of the CesT-extension motif in other highly translocated effectors, may contribute to differential cargo recognition by CesT.


Asunto(s)
Escherichia coli Enteropatógena , Proteínas de Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Dominios y Motivos de Interacción de Proteínas , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Células HeLa , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Organismos Modificados Genéticamente , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estructura Cuaternaria de Proteína , Transporte de Proteínas
9.
PLoS Pathog ; 14(4): e1007032, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29709025

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1005907.].

10.
Nature ; 510(7506): 503-6, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24965651

RESUMEN

The emergence and spread of carbapenem-resistant Gram-negative pathogens is a global public health problem. The acquisition of metallo-ß-lactamases (MBLs) such as NDM-1 is a principle contributor to the emergence of carbapenem-resistant Gram-negative pathogens that threatens the use of penicillin, cephalosporin and carbapenem antibiotics to treat infections. To date, a clinical inhibitor of MBLs that could reverse resistance and re-sensitize resistant Gram-negative pathogens to carbapenems has not been found. Here we have identified a fungal natural product, aspergillomarasmine A (AMA), that is a rapid and potent inhibitor of the NDM-1 enzyme and another clinically relevant MBL, VIM-2. AMA also fully restored the activity of meropenem against Enterobacteriaceae, Acinetobacter spp. and Pseudomonas spp. possessing either VIM or NDM-type alleles. In mice infected with NDM-1-expressing Klebsiella pneumoniae, AMA efficiently restored meropenem activity, demonstrating that a combination of AMA and a carbapenem antibiotic has therapeutic potential to address the clinical challenge of MBL-positive carbapenem-resistant Gram-negative pathogens.


Asunto(s)
Ácido Aspártico/análogos & derivados , Carbapenémicos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Tienamicinas/farmacología , Resistencia betalactámica/efectos de los fármacos , Inhibidores de beta-Lactamasas , Animales , Antibacterianos/farmacología , Ácido Aspártico/aislamiento & purificación , Ácido Aspártico/farmacología , Aspergillus/química , Productos Biológicos/aislamiento & purificación , Productos Biológicos/farmacología , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Bacterias Gramnegativas/enzimología , Bacterias Gramnegativas/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , Klebsiella pneumoniae/genética , Meropenem , Ratones , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
11.
PLoS Pathog ; 13(7): e1006497, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704543

RESUMEN

The evolution of bacterial pathogenicity, heavily influenced by horizontal gene transfer, provides new virulence factors and regulatory connections that alter bacterial phenotypes. Salmonella pathogenicity islands 1 and 2 (SPI-1 and SPI-2) are chromosomal regions that were acquired at different evolutionary times and are essential for Salmonella virulence. In the intestine of mammalian hosts, Salmonella expresses the SPI-1 genes that mediate its invasion to the gut epithelium. Once inside the cells, Salmonella down-regulates the SPI-1 genes and induces the expression of the SPI-2 genes, which favor its intracellular replication. The mechanism by which the invasion machinery is deactivated following successful invasion of host cells is not known. Here, we show that the SPI-2 encoded transcriptional regulator SsrB, which positively controls SPI-2, acts as a dual regulator that represses expression of SPI-1 during intracellular stages of infection. The mechanism of this SPI-1 repression by SsrB was direct and acts upon the hilD and hilA regulatory genes. The phenotypic effect of this molecular switch activity was a significant reduction in invasion ability of S. enterica serovar Typhimurium while promoting the expression of genes required for intracellular survival. During mouse infections, Salmonella mutants lacking SsrB had high levels of hilA (SPI-1) transcriptional activity whereas introducing a constitutively active SsrB led to significant hilA repression. Thus, our results reveal a novel SsrB-mediated mechanism of transcriptional crosstalk between SPI-1 and SPI-2 that helps Salmonella transition to the intracellular lifestyle.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Factores de Transcripción/metabolismo , Animales , Proteínas Bacterianas/genética , Islas Genómicas , Humanos , Ratones , Salmonella typhimurium/genética , Factores de Transcripción/genética , Virulencia
13.
PLoS Pathog ; 12(10): e1005907, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27711220

RESUMEN

Crohn's disease (CD) is a chronic inflammatory condition of diverse etiology. Exposure to foodborne pathogens causing acute gastroenteritis produces a long-term risk of CD well into the post-infectious period but the mechanistic basis for this ongoing relationship to disease onset is unknown. We developed two novel models to study the comorbidity of acute gastroenteritis caused by Salmonella Typhimurium or Citrobacter rodentium in mice colonized with adherent-invasive Escherichia coli (AIEC), a bacterial pathobiont linked to CD. Here, we show that disease activity in the post-infectious period after gastroenteritis is driven by the tissue-associated expansion of the resident AIEC pathobiont, with an attendant increase in immunopathology, barrier defects, and delays in mucosal restitution following pathogen clearance. These features required AIEC resistance to host defense peptides and a fulminant inflammatory response to the enteric pathogen. Our results suggest that individuals colonized by AIEC at the time of acute infectious gastroenteritis may be at greater risk for CD onset. Importantly, our data identify AIEC as a tractable disease modifier, a finding that could be exploited in the development of therapeutic interventions following infectious gastroenteritis in at-risk individuals.


Asunto(s)
Coinfección/complicaciones , Enfermedad de Crohn/microbiología , Gastroenteritis/complicaciones , Animales , Citrobacter rodentium , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/complicaciones , Escherichia coli , Infecciones por Escherichia coli/complicaciones , Femenino , Inmunohistoquímica , Inflamación/complicaciones , Inflamación/microbiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Salmonelosis Animal/complicaciones , Salmonella typhimurium
14.
PLoS Pathog ; 11(2): e1004648, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25643352

RESUMEN

Chemokines have been shown to be effective bactericidal molecules against a variety of bacteria and fungi in vitro. These direct antimicrobial effects are independent of their chemotactic activities involving immunological receptors. However, the direct biological role that these proteins may play in host defense, particularly against intestinal pathogens, is poorly understood. Here, we show that CXCL9, an ELR- chemokine, exhibits direct antimicrobial activity against Citrobacter rodentium, an attaching/effacing pathogen that infects the gut mucosa. Inhibition of this antimicrobial activity in vivo using anti-CXCL9 antibodies increases host susceptibility to C. rodentium infection with pronounced bacterial penetration into crypts, increased bacterial load, and worsened tissue pathology. Using Rag1(-/-) mice and CXCR3(-/-) mice, we demonstrate that the role for CXCL9 in protecting the gut mucosa is independent of an adaptive response or its immunological receptor, CXCR3. Finally, we provide evidence that phagocytes function in tandem with NK cells for robust CXCL9 responses to C. rodentium. These findings identify a novel role for the immune cell-derived CXCL9 chemokine in directing a protective antimicrobial response in the intestinal mucosa.


Asunto(s)
Quimiocina CXCL9/inmunología , Infecciones por Enterobacteriaceae/inmunología , Mucosa Intestinal/inmunología , Transducción de Señal/inmunología , Animales , Quimiocinas/inmunología , Citrobacter rodentium/inmunología , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Nature ; 478(7370): 506-10, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21993626

RESUMEN

Technological advances in DNA recovery and sequencing have drastically expanded the scope of genetic analyses of ancient specimens to the extent that full genomic investigations are now feasible and are quickly becoming standard. This trend has important implications for infectious disease research because genomic data from ancient microbes may help to elucidate mechanisms of pathogen evolution and adaptation for emerging and re-emerging infections. Here we report a reconstructed ancient genome of Yersinia pestis at 30-fold average coverage from Black Death victims securely dated to episodes of pestilence-associated mortality in London, England, 1348-1350. Genetic architecture and phylogenetic analysis indicate that the ancient organism is ancestral to most extant strains and sits very close to the ancestral node of all Y. pestis commonly associated with human infection. Temporal estimates suggest that the Black Death of 1347-1351 was the main historical event responsible for the introduction and widespread dissemination of the ancestor to all currently circulating Y. pestis strains pathogenic to humans, and further indicates that contemporary Y. pestis epidemics have their origins in the medieval era. Comparisons against modern genomes reveal no unique derived positions in the medieval organism, indicating that the perceived increased virulence of the disease during the Black Death may not have been due to bacterial phenotype. These findings support the notion that factors other than microbial genetics, such as environment, vector dynamics and host susceptibility, should be at the forefront of epidemiological discussions regarding emerging Y. pestis infections.


Asunto(s)
Genoma Bacteriano/genética , Peste/microbiología , Yersinia pestis/genética , Yersinia pestis/aislamiento & purificación , Cromosomas Bacterianos/genética , Mapeo Contig , Pulpa Dental/microbiología , Evolución Molecular , Historia Medieval , Humanos , Londres/epidemiología , Datos de Secuencia Molecular , Filogenia , Peste/epidemiología , Peste/transmisión , Plásmidos/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Virulencia/genética , Yersinia pestis/clasificación
16.
Mol Microbiol ; 95(4): 678-91, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25442048

RESUMEN

The two-component regulatory system SsrA-SsrB in Salmonella enterica controls expression of a virulence gene program required for intracellular survival in host cells. SsrA signaling is induced within the acidic host vacuole in which the bacteria reside; however, the mechanism by which SsrA senses this intracellular environment is unknown. Here, we show that the periplasmic sensor domain of SsrA is enriched in histidine residues that increase SsrA signaling below external pH of 6. While no single histidine accounted for the full acid-responsiveness of SsrA, we localized the acid-responsiveness principally to five histidines in the C-terminal end of the periplasmic sensor domain, with input from additional histidines in the N-terminal end of the senor. A sensor mutant lacking critical pH-responsive histidines was defective for acid-promoted activity, yet retained basal activity similar to wild type at neutral pH, indicating that the role of these histidines is to enhance signaling in response to acidification. In support of this, a pH-blind mutant was insensitive to the vacuole acidification blocking activity of bafilomycin, and was attenuated for competitive fitness during infection of mice. Our data demonstrate that SsrA contains a histidine-rich periplasmic sensor that enhances signaling in response to the innate host defense of vacuolar acidification.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Histidina/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Salmonella typhimurium/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Inhibidores Enzimáticos/farmacología , Aptitud Genética , Concentración de Iones de Hidrógeno , Macrólidos/farmacología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Periplasma , Estructura Terciaria de Proteína , Salmonelosis Animal/microbiología , Salmonella typhimurium/enzimología , Salmonella typhimurium/patogenicidad , Transducción de Señal , Vacuolas/metabolismo , Virulencia
17.
J Biol Chem ; 289(34): 23734-44, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25035427

RESUMEN

A number of Gram-negative pathogens utilize type III secretion systems (T3SSs) to inject bacterial effector proteins into the host. An important component of T3SSs is a conserved ATPase that captures chaperone-effector complexes and energizes their dissociation to facilitate effector translocation. To date, there has been limited work characterizing the chaperone-T3SS ATPase interaction despite it being a fundamental aspect of T3SS function. In this study, we present the 2.1 Å resolution crystal structure of the Salmonella enterica SPI-2-encoded ATPase, SsaN. Our structure revealed a local and functionally important novel feature in helix 10 that we used to define the interaction domain relevant to chaperone binding. We modeled the interaction between the multicargo chaperone, SrcA, and SsaN and validated this model using mutagenesis to identify the residues on both the chaperone and ATPase that mediate the interaction. Finally, we quantified the benefit of this molecular interaction on bacterial fitness in vivo using chromosomal exchange of wild-type ssaN with mutants that retain ATPase activity but no longer capture the chaperone. Our findings provide insight into chaperone recognition by T3SS ATPases and demonstrate the importance of the chaperone-T3SS ATPase interaction for the pathogenesis of Salmonella.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Sitios de Unión , Cristalización , Femenino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Virulencia
18.
Infect Immun ; 82(8): 3383-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866805

RESUMEN

Host defense peptides secreted by colonocytes and Paneth cells play a key role in innate host defenses in the gut. In Crohn's disease, the burden of tissue-associated Escherichia coli commonly increases at epithelial surfaces where host defense peptides concentrate, suggesting that this bacterial population might actively resist this mechanism of bacterial killing. Adherent-invasive E. coli (AIEC) is associated with Crohn's disease; however, the colonization determinants of AIEC in the inflamed gut are undefined. Here, we establish that host defense peptide resistance contributes to host colonization by Crohn's-associated AIEC. We identified a plasmid-encoded genomic island (called PI-6) in AIEC strain NRG857c that confers high-level resistance to α-helical cationic peptides and α- and ß-defensins. Deletion of PI-6 sensitized strain NRG857c to these host defense molecules, reduced its competitive fitness in a mouse model of infection, and attenuated its ability to induce cecal pathology. This phenotype is due to two genes in PI-6, arlA, which encodes a Mig-14 family protein implicated in defensin resistance, and arlC, an OmpT family outer membrane protease. Implicit in these findings are new bacterial targets whose inhibition might limit AIEC burden and disease in the gut.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Enfermedad de Crohn/microbiología , Farmacorresistencia Bacteriana , Proteínas de Escherichia coli/metabolismo , Escherichia coli/inmunología , Péptido Hidrolasas/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/genética , Femenino , Eliminación de Gen , Islas Genómicas , Humanos , Ratones , Péptido Hidrolasas/genética , Plásmidos , Factores de Virulencia/genética
19.
Eur J Immunol ; 43(10): 2638-49, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23775576

RESUMEN

Extracellular attaching and effacing (A/E) pathogens including pathogenic Escherichia coli colonize the host gut causing diarrhea and inflammation. Although much is known regarding the pathogenesis of A/E bacteria, there remains an incomplete understanding of host immune responses to these microbes. NK cells are an important source of IFN-γ and are essential for early innate responses to viral pathogens; however, their role during extracellular bacterial infections is still largely unexplored. We studied the host response to the murine A/E pathogen Citrobacter rodentium to investigate NK-cell function during infection. NK1.1⁺ cell depletions and analysis of colonic intestinal inflammation following Citrobacter infection demonstrated that CD3⁻NK1.1⁺ cells play an important role in the initial clearance of C. rodentium, as evidenced by higher bacterial load, intestinal pathology, and crypt hyperplasia at the peak of inflammation in depleted mice. Loss of CD3⁻NK1.1⁺ cells resulted in lower colonic IFN-γ, TNF-α, and IL-12, and a delay in homing of IFN-γ⁺CD4⁺ T cells to the gut. Loss of this response resulted in lower anti-C. rodentium IgG in NK1.1-depleted mice. These data establish that CD3⁻NK1.1⁺ cells are critical for inducing an early Th1 response involved in clearance of a pathogen that is restricted to the gastrointestinal tract.


Asunto(s)
Citrobacter rodentium/inmunología , Colon/inmunología , Infecciones por Enterobacteriaceae/inmunología , Escherichia coli/inmunología , Hiperplasia/inmunología , Células Asesinas Naturales/inmunología , Células TH1/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Ly/metabolismo , Carga Bacteriana/inmunología , Complejo CD3/metabolismo , Movimiento Celular/inmunología , Colon/microbiología , Colon/patología , Femenino , Hiperplasia/microbiología , Interferón gamma/genética , Interferón gamma/metabolismo , Activación de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo
20.
PLoS Pathog ; 8(6): e1002773, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761574

RESUMEN

Bacterial pathogens often manipulate host immune pathways to establish acute and chronic infection. Many Gram-negative bacteria do this by secreting effector proteins through a type III secretion system that alter the host response to the pathogen. In this study, we determined that the phage-encoded GogB effector protein in Salmonella targets the host SCF E3 type ubiquitin ligase through an interaction with Skp1 and the human F-box only 22 (FBXO22) protein. Domain mapping and functional knockdown studies indicated that GogB-containing bacteria inhibited IκB degradation and NFκB activation in macrophages, which required Skp1 and a eukaryotic-like F-box motif in the C-terminal domain of GogB. GogB-deficient Salmonella were unable to limit NFκB activation, which lead to increased proinflammatory responses in infected mice accompanied by extensive tissue damage and enhanced colonization in the gut during long-term chronic infections. We conclude that GogB is an anti-inflammatory effector that helps regulate inflammation-enhanced colonization by limiting tissue damage during infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas F-Box/metabolismo , Interacciones Huésped-Parásitos/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Infecciones por Salmonella/metabolismo , Animales , Proteínas Bacterianas/inmunología , Western Blotting , Proteínas F-Box/inmunología , Femenino , Técnicas de Silenciamiento del Gen , Transferencia de Gen Horizontal , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Citoplasmáticos y Nucleares/inmunología , Proteínas Quinasas Asociadas a Fase-S/inmunología , Infecciones por Salmonella/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA