Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Microbiol ; 24(1): 234, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951769

RESUMEN

BACKGROUND: Klebsiella aerogenes is an opportunistic pathogen that causes a wide variety of infections. Due to the rising problem of antibiotic resistance, novel antibiotics and strategies to combat bacterial infections are needed. Host-specific bacteriophages are natural enemies of bacteria and can be used in phage therapy as an alternative form of treatment against bacterial infections. Jumbo phages are defined as phages with genomes larger than 200 kb. Relatively few studies have been done on jumbo phages compared to smaller phages. RESULTS: A novel phage, fENko-Kae01, was isolated from a commercial phage cocktail. Genomic analysis revealed that fENko-Kae01 is a lytic jumbo phage with a 360 kb genome encoding 578 predicted genes. No highly similar phage genomes were identified and fENko-Kae01 may be a completely new genus representative. No known genes associated with lysogenic life cycle, bacterial virulence, or antibiotic resistance were identified. The phage had myovirus morphology and a narrow host range. Phage resistant bacterial mutants emerged under phage selection. Whole genome sequencing revealed that the biogenesis of the flagellum was affected in four mutants and the lack of functional flagellum was confirmed in motility assays. Furthermore, phage fENKo-Kae01 failed to adsorb on the non-motile mutants indicating that the bacterial flagellum is the phage-binding receptor. CONCLUSIONS: fENko-Kae01 is a novel jumbo bacteriophage that is considered safe for phage therapy. fENko-Kae01 uses the flagellum as the phage-binding receptor and may represent a completely novel genus.


Asunto(s)
Bacteriófagos , Enterobacter aerogenes , Flagelos , Genoma Viral , Especificidad del Huésped , Bacteriófagos/genética , Bacteriófagos/clasificación , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Flagelos/virología , Flagelos/genética , Enterobacter aerogenes/virología , Enterobacter aerogenes/genética , Secuenciación Completa del Genoma , Myoviridae/genética , Myoviridae/aislamiento & purificación , Myoviridae/clasificación , Myoviridae/fisiología
2.
Nucleic Acids Res ; 50(7): 3985-3997, 2022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35357498

RESUMEN

Yersinia phage YerA41 is morphologically similar to jumbo bacteriophages. The isolated genomic material of YerA41 could not be digested by restriction enzymes, and used as a template by conventional DNA polymerases. Nucleoside analysis of the YerA41 genomic material, carried out to find out whether this was due to modified nucleotides, revealed the presence of a ca 1 kDa substitution of thymidine with apparent oligosaccharide character. We identified and purified the phage DNA polymerase (DNAP) that could replicate the YerA41 genomic DNA even without added primers. Cryo-electron microscopy (EM) was used to characterize structural details of the phage particle. The storage capacity of the 131 nm diameter head was calculated to accommodate a significantly longer genome than that of the 145 577 bp genomic DNA of YerA41 determined here. Indeed, cryo-EM revealed, in contrast to the 25 Å in other phages, spacings of 33-36 Å between shells of the genomic material inside YerA41 heads suggesting that the heavily substituted thymidine increases significantly the spacing of the DNA packaged inside the capsid. In conclusion, YerA41 appears to be an unconventional phage that packages thymidine-modified genomic DNA into its capsids along with its own DNAP that has the ability to replicate the genome.


Asunto(s)
Bacteriófagos , Bacteriófagos/química , Bacteriófagos/genética , Cápside , Microscopía por Crioelectrón , ADN Viral/genética , ADN Polimerasa Dirigida por ADN/genética , Genoma Viral/genética , Timidina
3.
Clin Infect Dis ; 76(1): 103-112, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35676823

RESUMEN

BACKGROUND: Nontuberculous Mycobacterium infections, particularly Mycobacterium abscessus, are increasingly common among patients with cystic fibrosis and chronic bronchiectatic lung diseases. Treatment is challenging due to intrinsic antibiotic resistance. Bacteriophage therapy represents a potentially novel approach. Relatively few active lytic phages are available and there is great variation in phage susceptibilities among M. abscessus isolates, requiring personalized phage identification. METHODS: Mycobacterium isolates from 200 culture-positive patients with symptomatic disease were screened for phage susceptibilities. One or more lytic phages were identified for 55 isolates. Phages were administered intravenously, by aerosolization, or both to 20 patients on a compassionate use basis and patients were monitored for adverse reactions, clinical and microbiologic responses, the emergence of phage resistance, and phage neutralization in serum, sputum, or bronchoalveolar lavage fluid. RESULTS: No adverse reactions attributed to therapy were seen in any patient regardless of the pathogen, phages administered, or the route of delivery. Favorable clinical or microbiological responses were observed in 11 patients. Neutralizing antibodies were identified in serum after initiation of phage delivery intravenously in 8 patients, potentially contributing to lack of treatment response in 4 cases, but were not consistently associated with unfavorable responses in others. Eleven patients were treated with only a single phage, and no phage resistance was observed in any of these. CONCLUSIONS: Phage treatment of Mycobacterium infections is challenging due to the limited repertoire of therapeutically useful phages, but favorable clinical outcomes in patients lacking any other treatment options support continued development of adjunctive phage therapy for some mycobacterial infections.


Asunto(s)
Bacteriófagos , Fibrosis Quística , Infecciones por Mycobacterium no Tuberculosas , Mycobacterium , Terapia de Fagos , Humanos , Ensayos de Uso Compasivo , Preparaciones Farmacéuticas , Infecciones por Mycobacterium no Tuberculosas/microbiología , Fibrosis Quística/microbiología , Antibacterianos/uso terapéutico
4.
Arch Virol ; 168(2): 44, 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36609878

RESUMEN

The rise of antibiotic resistance in bacterial strains has led to vigorous exploration for alternative treatments. To this end, phage therapy has been revisited, and it is gaining increasing attention, as it may represent an efficient alternative for treating multiresistant pathogenic bacteria. Phage therapy is considered safe, and phages do not infect eukaryotic cells. There have been many studies investigating phage-host bacteria interactions and the ability of phages to target specific hosts. Escherichia coli is the causative agent of a multitude of infections, ranging from urinary tract infections to sepsis, with growing antibiotic resistance. In this study, we characterized the Escherichia phage fBC-Eco01, which was isolated from a water sample collected at Oued, Tunis. Electron microscopy showed that fBC-Eco01 phage particles have siphovirus morphology, with an icosahedral head of 61 ± 3 nm in diameter and a non-contractile tail of 94 ± 2 nm in length and 12 ± 0.9 nm in width. The genome of fBC-Eco01 is a linear double-stranded DNA of 43.466 bp with a GC content of 50.4%. Comparison to databases allowed annotation of the functions to 39 of the 78 predicted gene products. A single-step growth curve revealed that fBC-Eco01 has a latent period of 30 minutes and a burst size of 175 plaque-forming units (PFU) per infected cell. Genomic analysis indicated that fBC-Eco01 is a member of the subfamily Guernseyvirinae. It is most closely related to a group of phages of the genus Kagunavirus that infect Enterobacter, Raoultella, and Escherichia strains.


Asunto(s)
Bacteriófagos , Siphoviridae , Aguas Residuales , Túnez , Genoma Viral , Bacteriófagos/genética , Escherichia coli/genética , Siphoviridae/genética
5.
Arch Virol ; 168(9): 228, 2023 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-37574509

RESUMEN

Acinetobacter baumannii is an opportunistic pathogen that is mostly associated with hospital-acquired infections. The rapid emergence of multi- and pan-drug-resistant Acinetobacter strains poses an increasing challenge in hospitals. Phage therapy offers one treatment option for infections caused by A. baumannii. We isolated three phages from Beninese hospital wastewater - fBenAci001, fBenAci002, and fBenAci003 - that infected clinical A. baumannii strains from Finnish patients. Phylogenetic analysis showed that these phages resemble phages of the genus Friunavirus, family Autographiviridae. The isolated phages meet the requirements set for phages used for phage therapy. However, they were found to have a narrow host range, which may limit their therapeutic use.


Asunto(s)
Acinetobacter baumannii , Bacteriófagos , Humanos , Bacteriófagos/genética , Aguas Residuales , Filogenia , Especificidad del Huésped , Antibacterianos
6.
Arch Virol ; 167(5): 1333-1341, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35399144

RESUMEN

Characterization of bacteriophages facilitates better understanding of their biology, host specificity, genomic diversity, and adaptation to their bacterial hosts. This, in turn, is important for the exploitation of phages for therapeutic purposes, as the use of uncharacterized phages may lead to treatment failure. The present study describes the isolation and characterization of a bacteriophage effective against the important clinical pathogen Escherichia coli, which shows increasing accumulation of antibiotic resistance. Phage fEg-Eco19, which is specific for a clinical E. coli strain, was isolated from an Egyptian sewage sample. Phage fEg-Eco19 formed clear, sharp-edged, round plaques. Electron microscopy showed that the isolated phage is tailed and therefore belongs to the order Caudovirales, and morphologically, it resembles siphoviruses. The diameter of the icosahedral head of fEg-Eco19 is 68 ± 2 nm, and the non-contractile tail length and diameter are 118 ± 0.2 and 13 ± 0.6 nm, respectively. The host range of the phage was found to be narrow, as it infected only two out of 137 clinical E. coli strains tested. The phage genome is 45,805 bp in length with a GC content of 50.3% and contains 76 predicted genes. Comparison of predicted and experimental restriction digestion patterns allowed rough mapping of the physical ends of the phage genome, which was confirmed using the PhageTerm tool. Annotation of the predicted genes revealed gene products belonging to several functional groups, including regulatory proteins, DNA packaging and phage structural proteins, host lysis proteins, and proteins involved in DNA/RNA metabolism and replication.


Asunto(s)
Bacteriófagos , Caudovirales , Antibacterianos/farmacología , Bacteriófagos/genética , Caudovirales/genética , Escherichia coli/genética , Genoma Viral , Especificidad del Huésped
7.
Microb Pathog ; 141: 103993, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31988008

RESUMEN

Yersinia pestis, a Gram-negative bacterium, is the etiologic agent of plague. A hallmark of Y. pestis infection is the organism's ability to rapidly disseminate through an animal host. Y. pestis expresses the outer membrane protein, Ail (Attachment invasion locus), which is associated with host invasion and serum resistance. However, whether Ail plays a role in host dissemination remains unclear. In this study, C57BL/6J mice were challenged with a defined Y. pestis strain, KimD27, or an isogenic ail-deleted mutant derived from KimD27 via metacarpal paw pad inoculation, nasal drops, orogastric infection, or tail vein injection to mimic bubonic, pneumonic, oral, or septicemic plague, respectively. Our results showed that ail-deleted Y. pestis KimD27 lost the ability to invade host cells, leading to failed host dissemination in the pneumonic and oral plague models but not in the bubonic or septicemic plague models, which do not require invasiveness. Therefore, this study demonstrated that whether Ail plays a role in Y. pestis pathogenesis depends on the infection route.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Peste/microbiología , Factores de Virulencia/metabolismo , Virulencia , Yersinia pestis , Animales , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Boca/microbiología , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidad
8.
Infect Immun ; 87(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30348825

RESUMEN

Yersinia pseudotuberculosis is a Gram-negative enteropathogen and causes gastrointestinal infections. It disseminates from gut to mesenteric lymph nodes (MLNs), spleen, and liver of infected humans and animals. Although the molecular mechanisms for dissemination and infection are unclear, many Gram-negative enteropathogens presumably invade the small intestine via Peyer's patches to initiate dissemination. In this study, we demonstrate that Y. pseudotuberculosis utilizes its lipopolysaccharide (LPS) core to interact with CD209 receptors, leading to invasion of human dendritic cells (DCs) and murine macrophages. These Y. pseudotuberculosis-CD209 interactions result in bacterial dissemination to MLNs, spleens, and livers of both wild-type and Peyer's patch-deficient mice. The blocking of the Y. pseudotuberculosis-CD209 interactions by expression of O-antigen and with oligosaccharides reduces infectivity. Based on the well-documented studies in which HIV-CD209 interaction leads to viral dissemination, we therefore propose an infection route for Y. pseudotuberculosis where this pathogen, after penetrating the intestinal mucosal membrane, hijacks the Y. pseudotuberculosis-CD209 interaction antigen-presenting cells to reach their target destinations, MLNs, spleens, and livers.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/microbiología , Endocitosis , Interacciones Huésped-Patógeno , Lectinas Tipo C/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/microbiología , Receptores de Superficie Celular/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Animales , Adhesión Bacteriana , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica , Yersiniosis/microbiología , Yersiniosis/patología , Yersiniosis/fisiopatología
9.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31085704

RESUMEN

Salmonella enterica serovar Typhimurium, a Gram-negative bacterium, can cause infectious diseases ranging from gastroenteritis to systemic dissemination and infection. However, the molecular mechanisms underlying this bacterial dissemination have yet to be elucidated. A study indicated that using the lipopolysaccharide (LPS) core as a ligand, S Typhimurium was able to bind human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (hCD209a), an HIV receptor that promotes viral dissemination by hijacking antigen-presenting cells (APCs). In this study, we showed that S Typhimurium interacted with CD209s, leading to the invasion of APCs and potentially the dissemination to regional lymph nodes, spleen, and liver in mice. Shielding of the exposed LPS core through the expression of O-antigen reduces dissemination and infection. Thus, we propose that similar to HIV, S Typhimurium may also utilize APCs via interactions with CD209s as a way to disseminate to the lymph nodes, spleen, and liver to initiate host infection.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Lectinas Tipo C/fisiología , Receptores de Superficie Celular/fisiología , Salmonella typhimurium/patogenicidad , Animales , Células Presentadoras de Antígenos/microbiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Lipopolisacáridos/fisiología , Mananos/farmacología , Ratones , Ratones Endogámicos C57BL , Antígenos O/fisiología , Ganglios Linfáticos Agregados/fisiología , Fagocitosis , Células RAW 264.7
10.
Blood ; 130(3): 245-257, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28465342

RESUMEN

The iron-regulatory hormone hepcidin is induced early in infection, causing iron sequestration in macrophages and decreased plasma iron; this is proposed to limit the replication of extracellular microbes, but could also promote infection with macrophage-tropic pathogens. The mechanisms by which hepcidin and hypoferremia modulate host defense, and the spectrum of microbes affected, are poorly understood. Using mouse models, we show that hepcidin was selectively protective against siderophilic extracellular pathogens (Yersinia enterocolitica O9) by controlling non-transferrin-bound iron (NTBI) rather than iron-transferrin concentration. NTBI promoted the rapid growth of siderophilic but not nonsiderophilic bacteria in mice with either genetic or iatrogenic iron overload and in human plasma. Hepcidin or iron loading did not affect other key components of innate immunity, did not indiscriminately promote intracellular infections (Mycobacterium tuberculosis), and had no effect on extracellular nonsiderophilic Y enterocolitica O8 or Staphylococcus aureus Hepcidin analogs may be useful for treatment of siderophilic infections.


Asunto(s)
Infecciones Relacionadas con Catéteres/inmunología , Hemocromatosis/inmunología , Hepcidinas/inmunología , Sobrecarga de Hierro/inmunología , Hierro/metabolismo , Infecciones Estafilocócicas/inmunología , Animales , Unión Competitiva , Infecciones Relacionadas con Catéteres/metabolismo , Infecciones Relacionadas con Catéteres/microbiología , Infecciones Relacionadas con Catéteres/mortalidad , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Expresión Génica , Hemocromatosis/metabolismo , Hemocromatosis/microbiología , Hemocromatosis/mortalidad , Hepcidinas/agonistas , Hepcidinas/deficiencia , Hepcidinas/genética , Humanos , Hierro/inmunología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/microbiología , Sobrecarga de Hierro/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Oligopéptidos/farmacología , Unión Proteica , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/mortalidad , Staphylococcus aureus , Análisis de Supervivencia , Transferrina/genética , Transferrina/metabolismo , Yersinia enterocolitica/efectos de los fármacos , Yersinia enterocolitica/crecimiento & desarrollo , Yersinia enterocolitica/metabolismo
11.
Arch Virol ; 164(8): 2171-2173, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31093759

RESUMEN

We report here the annotation of the complete genomes of four novel lytic Staphylococcus phages; Stab20, Stab21, Stab22 and Stab23. These phages have double-stranded DNA genomes ranging between 153,338 and 155,962 bp in size with terminal repeats of 10,814-12,304 bp. The genome analysis suggests that they represent new phage species within the genus Kayvirus in the subfamily Twortvirinae of the family Herelleviridae.


Asunto(s)
Myoviridae/genética , Staphylococcus/genética , ADN Viral/genética , Genoma Viral/genética , Genómica/métodos , Filogenia , Análisis de Secuencia de ADN/métodos , Fagos de Staphylococcus/genética
12.
Arch Virol ; 164(8): 2197-2199, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31123962

RESUMEN

We present here the isolation and characterization of Acinetobacter pittii phage vB_ApiM_fHyAci03 (fHyAci03), which belongs to the family Myoviridae. The fHyAci03 genome was found to be 165,975 bp in length and predicted to contain 255 genes. While the whole genome was 92.4% identical to Acinetobacter baumannii phage KARL-1, phylogenetic analysis based on phage long distal tail fiber amino acid sequences assigned fHyAci03 and KARL-1 to different subclusters, reflecting their different host species. Together with phylogenetic analysis, genome comparisons indicated that fHyAci03 is a novel member of the subfamily Tevenvirinae. Host range experiments revealed that fHyAci03 could infect two clinical strains of Acinetobacter nosocomialis and six clinical strains of A. pittii. Thus, fHyAci03 is a novel lytic phage that infects clinical Acinetobacter strains and represents a potential new candidate to be used in phage therapy.


Asunto(s)
Acinetobacter/virología , Bacteriófagos/genética , ADN Viral/genética , Genoma Viral/genética , Especificidad del Huésped/genética , Myoviridae/genética , Filogenia
13.
Mol Microbiol ; 103(6): 1065-1091, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28010054

RESUMEN

In bacteria, the RNA chaperone Hfq enables pairing of small regulatory RNAs with their target mRNAs and therefore is a key player of post-transcriptional regulation network. As a global regulator, Hfq is engaged in the adaptation to external environment, regulation of metabolism and bacterial virulence. In this study we used RNA-sequencing and quantitative proteomics (LC-MS/MS) to elucidate the role of this chaperone in the physiology and virulence of Yersinia enterocolitica serotype O:3. This global approach revealed the profound impact of Hfq on gene and protein expression. Furthermore, the role of Hfq in the cell morphology, metabolism, cell wall integrity, resistance to external stresses and pathogenicity was evaluated. Importantly, our results revealed that several alterations typical for the hfq-negative phenotype were due to derepression of the transcriptional factor RovM. The overexpression of RovM caused by the loss of Hfq chaperone resulted in extended growth defect, alterations in the lipid A structure, motility and biofilm formation defects, as well as changes in mannitol utilization. Furthermore, in Y. enterocolitica RovM only in the presence of Hfq affected the abundance of RpoS. Finally, the impact of hfq and rovM mutations on the virulence was assessed in the mouse infection model.


Asunto(s)
Proteínas Bacterianas/genética , Proteína de Factor 1 del Huésped/genética , Chaperonas Moleculares/genética , ARN Largo no Codificante/genética , ARN Pequeño no Traducido/genética , Secuencias Reguladoras de Ácido Ribonucleico/genética , Factor sigma/metabolismo , Factores de Transcripción/genética , Yersinia enterocolitica/genética , Yersinia enterocolitica/patogenicidad , Animales , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Biopelículas/crecimiento & desarrollo , Pared Celular/genética , Gastroenteritis/microbiología , Regulación Bacteriana de la Expresión Génica , Proteína de Factor 1 del Huésped/metabolismo , Lípido A/metabolismo , Manitol/metabolismo , Espectrometría de Masas , Ratones , Chaperonas Moleculares/metabolismo , Proteoma/genética , Proteómica , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo , Yersiniosis/microbiología , Yersinia enterocolitica/clasificación
14.
J Bacteriol ; 199(18)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28652301

RESUMEN

The second messenger cyclic dimeric GMP (c-di-GMP) is almost ubiquitous among bacteria as are the c-di-GMP turnover proteins, which mediate the transition between motility and sessility. EAL domain proteins have been characterized as c-di-GMP-specific phosphodiesterases. While most EAL domain proteins contain additional, usually N-terminal, domains, there is a distinct family of proteins with stand-alone EAL domains, exemplified by Salmonella enterica serovar Typhimurium proteins STM3611 (YhjH/PdeH), a c-di-GMP-specific phosphodiesterase, and the enzymatically inactive STM1344 (YdiV/CdgR) and STM1697, which regulate bacterial motility through interaction with the flagellar master regulator, FlhDC. We have analyzed the phylogenetic distribution of EAL-only proteins and their potential functions. Genes encoding EAL-only proteins were found in various bacterial phyla, although most of them were seen in proteobacteria, particularly enterobacteria. Based on the conservation of the active site residues, nearly all stand-alone EAL domains encoded by genomes from phyla other than proteobacteria appear to represent functional phosphodiesterases. Within enterobacteria, EAL-only proteins were found to cluster either with YhjH or with one of the subfamilies of YdiV-related proteins. EAL-only proteins from Shigella flexneri, Klebsiella pneumoniae, and Yersinia enterocolitica were tested for their ability to regulate swimming and swarming motility and formation of the red, dry, and rough (rdar) biofilm morphotype. In these tests, YhjH-related proteins S4210, KPN_01159, KPN_03274, and YE4063 displayed properties typical of enzymatically active phosphodiesterases, whereas S1641 and YE1324 behaved like members of the YdiV/STM1697 subfamily, with Yersinia enterocolitica protein YE1324 shown to downregulate motility in its native host. Of two closely related EAL-only proteins, YE2225 is an active phosphodiesterase, while YE1324 appears to interact with FlhD. These results suggest that in FlhDC-harboring beta- and gammaproteobacteria, some EAL-only proteins evolved to become catalytically inactive and regulate motility and biofilm formation by interacting with FlhDC.IMPORTANCE The EAL domain superfamily consists mainly of proteins with cyclic dimeric GMP-specific phosphodiesterase activity, but individual domains have been classified in three classes according to their functions and conserved amino acid signatures. Proteins that consist solely of stand-alone EAL domains cannot rely on other domains to form catalytically active dimers, and most of them fall into one of two distinct classes: catalytically active phosphodiesterases with well-conserved residues of the active site and the dimerization loop, and catalytically inactive YdiV/CdgR-like proteins that regulate bacterial motility by binding to the flagellar master regulator, FlhDC, and are found primarily in enterobacteria. The presence of apparently inactive EAL-only proteins in the bacteria that do not express FlhD suggests the existence of additional EAL interaction partners.


Asunto(s)
Biopelículas/crecimiento & desarrollo , GMP Cíclico/análogos & derivados , Enterobacteriaceae/genética , Enterobacteriaceae/fisiología , Regulación Bacteriana de la Expresión Génica , Locomoción , Hidrolasas Diéster Fosfóricas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Análisis por Conglomerados , Biología Computacional , Secuencia Conservada , GMP Cíclico/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Filogenia , Homología de Secuencia de Aminoácido
15.
Immunol Cell Biol ; 95(8): 666-675, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28405017

RESUMEN

Both complement activation and certain infections (including those with Yersinia sp.) may contribute to the pathogenesis of juvenile idiopathic arthritis (JIA). We investigated factors specific for the lectin pathway of complement: mannose-binding lectin (MBL), ficolins and MBL-associated serine protease-2 (MASP-2), in 144 patients and 98 controls. One hundred and six patients had oligoarticular disease and 38 had polyarticular disease. In 51 patients (out of 133 tested), Yersinia-reactive antibodies were found (JIA Ye+ group). MBL deficiency was significantly more frequent in the JIA Ye+ group than in patients without Yersinia-reactive antibodies or in controls. Median serum ficolin-2 level was significantly lower (and proportion of values deemed ficolin-2 insufficient greater) in JIA patients irrespective of their Yersinia antibody status. The minority (C) allele at -64 of the FCN2 gene was less frequent among JIA patients than among control subjects. No differences were found in the frequency of FCN3 gene +1637delC or MASP2 +359 A>G mutations nor for median values of serum ficolin-1, ficolin-3 or MASP-2. However, high levels of serum ficolin-3 were under-represented in patients, in contrast to MBL. MBL, ficolin-1, ficolin-2, ficolin-3 and MASP-2 were also readily detectable in synovial fluid samples but at a considerably lower level than in serum. Our findings suggest a possible role for the lectin pathway in the pathogenesis of JIA, perhaps secondary to a role in host defence, and indicate that investigations on the specificity of lectin pathway recognition molecules towards specific infectious agents in JIA might be fruitful.


Asunto(s)
Artritis Juvenil/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/genética , Lectinas/genética , Lectina de Unión a Manosa/genética , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Yersinia pseudotuberculosis/inmunología , Adolescente , Anticuerpos Antibacterianos/sangre , Artritis Juvenil/epidemiología , Niño , Preescolar , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Glicoproteínas/genética , Humanos , Lactante , Masculino , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Polimorfismo Genético , Yersiniosis/epidemiología , Ficolinas
16.
Food Microbiol ; 65: 36-43, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28400017

RESUMEN

To study the role of each two-component system (TCS) histidine kinase (HK) in stress tolerance of Listeria monocytogenes EGD-e, we monitored the growth of individual HK deletion mutant strains under heat (42.5 °C), acid (pH 5.6), alkali (pH 9.4), osmotic (6% NaCl), ethanol (3.5 vol%), and oxidative (5 mM H2O2) stresses. The growth of ΔliaS (Δlmo1021) strain was impaired under each stress, with the most notable decrease under heat and osmotic stresses. The ΔvirS (Δlmo1741) strain showed nearly completely restricted growth at high temperature and impaired growth in ethanol. The growth of ΔagrC (Δlmo0050) strain was impaired under osmotic stress and slightly under oxidative stress. We successfully complemented the HK mutations using a novel allelic exchange based approach. This approach avoided the copy-number problems associated with in trans complementation from a plasmid. The mutant phenotypes were restored to the wild-type level in the complemented strains. This study reveals novel knowledge on the HKs needed for growth of L. monocytogenes EGD-e under abovementioned stress conditions, with LiaS playing multiple roles in stress tolerance of L. monocytogenes EGD-e.


Asunto(s)
Álcalis/metabolismo , Etanol/metabolismo , Histidina Quinasa/metabolismo , Calor , Listeria monocytogenes/enzimología , Listeria monocytogenes/crecimiento & desarrollo , Estrés Oxidativo , Adaptación Fisiológica/genética , Dosificación de Gen , Prueba de Complementación Genética , Concentración de Iones de Hidrógeno , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Mutación , Fenotipo , Proteínas Quinasas/metabolismo , Eliminación de Secuencia
17.
Proc Natl Acad Sci U S A ; 111(18): 6768-73, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753568

RESUMEN

The genus Yersinia has been used as a model system to study pathogen evolution. Using whole-genome sequencing of all Yersinia species, we delineate the gene complement of the whole genus and define patterns of virulence evolution. Multiple distinct ecological specializations appear to have split pathogenic strains from environmental, nonpathogenic lineages. This split demonstrates that contrary to hypotheses that all pathogenic Yersinia species share a recent common pathogenic ancestor, they have evolved independently but followed parallel evolutionary paths in acquiring the same virulence determinants as well as becoming progressively more limited metabolically. Shared virulence determinants are limited to the virulence plasmid pYV and the attachment invasion locus ail. These acquisitions, together with genomic variations in metabolic pathways, have resulted in the parallel emergence of related pathogens displaying an increasingly specialized lifestyle with a spectrum of virulence potential, an emerging theme in the evolution of other important human pathogens.


Asunto(s)
Evolución Molecular , Virulencia/genética , Yersinia/genética , Yersinia/patogenicidad , Genoma Bacteriano , Humanos , Redes y Vías Metabólicas/genética , Filogenia , Especificidad de la Especie , Yersinia/metabolismo , Yersinia enterocolitica/genética , Yersinia enterocolitica/metabolismo , Yersinia enterocolitica/patogenicidad
18.
Appl Environ Microbiol ; 82(17): 5340-53, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27342557

RESUMEN

UNLABELLED: Bacteriophages present huge potential both as a resource for developing novel tools for bacterial diagnostics and for use in phage therapy. This potential is also valid for bacteriophages specific for Yersinia enterocolitica To increase our knowledge of Y. enterocolitica-specific phages, we characterized two novel yersiniophages. The genomes of the bacteriophages vB_YenM_TG1 (TG1) and vB_YenM_ϕR1-RT (ϕR1-RT), isolated from pig manure in Canada and from sewage in Finland, consist of linear double-stranded DNA of 162,101 and 168,809 bp, respectively. Their genomes comprise 262 putative coding sequences and 4 tRNA genes and share 91% overall nucleotide identity. Based on phylogenetic analyses of their whole-genome sequences and large terminase subunit protein sequences, a genus named Tg1virus within the family Myoviridae is proposed, with TG1 and ϕR1-RT (R1RT in the ICTV database) as member species. These bacteriophages exhibit a host range restricted to Y. enterocolitica and display lytic activity against the epidemiologically significant serotypes O:3, O:5,27, and O:9 at and below 25°C. Adsorption analyses of lipopolysaccharide (LPS) and OmpF mutants demonstrate that these phages use both the LPS inner core heptosyl residues and the outer membrane protein OmpF as phage receptors. Based on RNA sequencing and quantitative proteomics, we also demonstrate that temperature-dependent infection is due to strong repression of OmpF at 37°C. In addition, ϕR1-RT was shown to be able to enter into a pseudolysogenic state. Together, this work provides further insight into phage-host cell interactions by highlighting the importance of understanding underlying factors which may affect the abundance of phage host receptors on the cell surface. IMPORTANCE: Only a small number of bacteriophages infecting Y. enterocolitica, the predominant causative agent of yersiniosis, have been previously described. Here, two newly isolated Y. enterocolitica phages were studied in detail, with the aim of elucidating the host cell receptors required for infection. Our research further expands the repertoire of phages available for consideration as potential antimicrobial agents or as diagnostic tools for this important bacterial pathogen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/fisiología , Especificidad del Huésped , Porinas/metabolismo , Receptores Virales/metabolismo , Yersinia enterocolitica/virología , Proteínas Bacterianas/genética , Bacteriófagos/clasificación , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Genoma Viral , Humanos , Filogenia , Porinas/genética , Receptores Virales/genética , Temperatura , Replicación Viral , Yersiniosis/microbiología , Yersinia enterocolitica/genética , Yersinia enterocolitica/metabolismo
19.
Adv Exp Med Biol ; 918: 361-375, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27722870

RESUMEN

Bacteriophage play many varied roles in microbial ecology and evolution. This chapter collates a vast body of knowledge and expertise on Yersinia pestis phages, including the history of their isolation and classical methods for their isolation and identification. The genomic diversity of Y. pestis phage and bacteriophage islands in the Y. pestis genome are also discussed because all phage research represents a branch of genetics. In addition, our knowledge of the receptors that are recognized by Y. pestis phage, advances in phage therapy for Y. pestis infections, the application of phage in the detection of Y. pestis, and clustered regularly interspaced short palindromic repeats (CRISPRs) sequences of Y. pestis from prophage DNA are all reviewed here.


Asunto(s)
Bacteriófagos , Yersinia pestis/virología , Animales , Bacteriófagos/clasificación , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Variación Genética , Islas Genómicas , Humanos , Profagos/clasificación , Profagos/genética , Profagos/aislamiento & purificación , Receptores Virales/fisiología
20.
Duodecim ; 132(8): 712-9, 2016.
Artículo en Fi | MEDLINE | ID: mdl-27244930

RESUMEN

Antibiotic resistance of bacterial pathogens has increased, and new therapies are urgently needed. Bacteriophages (phages), viruses infecting and killing bacteria, are the most abundant organisms on earth. In nature there are several specific phages for every bacterium, controlling bacterial numbers and maintaining ecological balance. Phage therapy, i.e., treating bacterial infections with phages, offers an alternative complementary to antibiotics as phages infect and kill also multi-resistant bacteria. Phages possess narrow host specificity, each phage infecting only a few bacterial species or strains. Thereby phages do not harm the normal microbiota as antibiotics do. We aim to begin clinical trials of phage therapy in Finland.


Asunto(s)
Infecciones Bacterianas/terapia , Bacteriófagos , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Finlandia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA