Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Commun ; 14(1): 4196, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37452045

RESUMEN

The ever-growing rise of antibiotic resistance among bacterial pathogens is one of the top healthcare threats today. Although combination antibiotic therapies represent a potential approach to more efficiently combat infections caused by susceptible and drug-resistant bacteria, only a few known drug pairs exhibit synergy/cooperativity in killing bacteria. Here, we discover that well-known ribosomal antibiotics, hygromycin A (HygA) and macrolides, which target peptidyl transferase center and peptide exit tunnel, respectively, can act cooperatively against susceptible and drug-resistant bacteria. Remarkably, HygA slows down macrolide dissociation from the ribosome by 60-fold and enhances the otherwise weak antimicrobial activity of the newest-generation macrolide drugs known as ketolides against macrolide-resistant bacteria. By determining a set of high-resolution X-ray crystal structures of drug-sensitive wild-type and macrolide-resistant Erm-methylated 70S ribosomes in complex with three HygA-macrolide pairs, we provide a structural rationale for the binding cooperativity of these drugs and also uncover the molecular mechanism of overcoming Erm-type resistance by macrolides acting together with hygromycin A. Altogether our structural, biochemical, and microbiological findings lay the foundation for the subsequent development of synergistic antibiotic tandems with improved bactericidal properties against drug-resistant pathogens, including those expressing erm genes.


Asunto(s)
Cetólidos , Macrólidos , Macrólidos/farmacología , Antibacterianos/química , Cinamatos/farmacología , Higromicina B/farmacología , Cetólidos/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Bacterias/metabolismo , Farmacorresistencia Bacteriana/genética
2.
Cell ; 184(21): 5405-5418.e16, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34619078

RESUMEN

Lyme disease is on the rise. Caused by a spirochete Borreliella burgdorferi, it affects an estimated 500,000 people in the United States alone. The antibiotics currently used to treat Lyme disease are broad spectrum, damage the microbiome, and select for resistance in non-target bacteria. We therefore sought to identify a compound acting selectively against B. burgdorferi. A screen of soil micro-organisms revealed a compound highly selective against spirochetes, including B. burgdorferi. Unexpectedly, this compound was determined to be hygromycin A, a known antimicrobial produced by Streptomyces hygroscopicus. Hygromycin A targets the ribosomes and is taken up by B. burgdorferi, explaining its selectivity. Hygromycin A cleared the B. burgdorferi infection in mice, including animals that ingested the compound in a bait, and was less disruptive to the fecal microbiome than clinically relevant antibiotics. This selective antibiotic holds the promise of providing a better therapeutic for Lyme disease and eradicating it in the environment.


Asunto(s)
Antibacterianos/uso terapéutico , Enfermedad de Lyme/tratamiento farmacológico , Animales , Borrelia burgdorferi/efectos de los fármacos , Calibración , Cinamatos/química , Cinamatos/farmacología , Cinamatos/uso terapéutico , Evaluación Preclínica de Medicamentos , Heces/microbiología , Femenino , Células HEK293 , Células Hep G2 , Humanos , Higromicina B/análogos & derivados , Higromicina B/química , Higromicina B/farmacología , Higromicina B/uso terapéutico , Enfermedad de Lyme/microbiología , Ratones , Pruebas de Sensibilidad Microbiana , Microbiota/efectos de los fármacos
3.
mBio ; 11(2)2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32291298

RESUMEN

Staphylococcus aureus is a major concern in human health care, mostly due to the increasing prevalence of antibiotic resistance. Intracellular localization of S. aureus plays a key role in recurrent infections by protecting the pathogens from antibiotics and immune responses. Peptidoglycan hydrolases (PGHs) are highly specific bactericidal enzymes active against both drug-sensitive and -resistant bacteria. However, PGHs able to effectively target intracellular S. aureus are not yet available. To overcome this limitation, we first screened 322 recombineered PGHs for staphylolytic activity under conditions found inside eukaryotic intracellular compartments. The most active constructs were modified by fusion to different cell-penetrating peptides (CPPs), resulting in increased uptake and enhanced intracellular killing (reduction by up to 4.5 log units) of various S. aureus strains (including methicillin-resistant S. aureus [MRSA]) in different tissue culture infection models. The combined application of synergistic PGH-CPP constructs further enhanced their intracellular efficacy. Finally, synergistically active PGH-CPP cocktails reduced the total S. aureus by more than 2.2 log units in a murine abscess model after peripheral injection. Significantly more intracellular bacteria were killed by the PGH-CPPs than by the PGHs alone. Collectively, our findings show that CPP-fused PGHs are effective novel protein therapeutics against both intracellular and drug-resistant S. aureusIMPORTANCE The increasing prevalence of antibiotic-resistant bacteria is one of the most urgent problems of our time. Staphylococcus aureus is an important human pathogen that has acquired several mechanisms to evade antibiotic treatment. In addition, S. aureus is able to invade and persist within human cells, hiding from the immune response and antibiotic therapies. For these reasons, novel antibacterial strategies against these pathogens are needed. Here, we developed lytic enzymes which are able to effectively target drug-resistant and intracellular S. aureus Fusion of these so-called enzybiotics to cell-penetrating peptides enhanced their uptake and intracellular bactericidal activity in cell culture and in an abscess mouse model. Our results suggest that cell-penetrating enzybiotics are a promising new class of therapeutics against staphylococcal infections.


Asunto(s)
Antibacterianos/uso terapéutico , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/uso terapéutico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Células 3T3-L1 , Células A549 , Absceso/tratamiento farmacológico , Absceso/microbiología , Animales , Antibacterianos/química , Farmacorresistencia Bacteriana , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/uso terapéutico
4.
Nat Commun ; 10(1): 1149, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850614

RESUMEN

Treatment failure in biofilm-associated bacterial infections is an important healthcare issue. In vitro studies and mouse models suggest that bacteria enter a slow-growing/non-growing state that results in transient tolerance to antibiotics in the absence of a specific resistance mechanism. However, little clinical confirmation of antibiotic tolerant bacteria in patients exists. In this study we investigate a Staphylococcus epidermidis pacemaker-associated endocarditis, in a patient who developed a break-through bacteremia despite taking antibiotics to which the S. epidermidis isolate is fully susceptible in vitro. Characterization of the clinical S. epidermidis isolates reveals in-host evolution over the 16-week infection period, resulting in increased antibiotic tolerance of the entire population due to a prolonged lag time until growth resumption and a reduced growth rate. Furthermore, we observe adaptation towards an increased biofilm formation capacity and genetic diversification of the S. epidermidis isolates within the patient.


Asunto(s)
Antibacterianos/farmacología , Bacteriemia/microbiología , Resistencia a Múltiples Medicamentos/genética , Endocarditis/microbiología , Interacciones Huésped-Patógeno/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/genética , Adulto , Bacteriemia/tratamiento farmacológico , Bacteriemia/patología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Tolerancia a Medicamentos/genética , Endocarditis/tratamiento farmacológico , Endocarditis/patología , Evolución Molecular , Fluoroquinolonas/farmacología , Glicopéptidos/farmacología , Humanos , Mutación INDEL , Masculino , Pruebas de Sensibilidad Microbiana , Marcapaso Artificial/microbiología , Péptidos Cíclicos/farmacología , Filogenia , Polimorfismo de Nucleótido Simple , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/crecimiento & desarrollo , Staphylococcus epidermidis/aislamiento & purificación , beta-Lactamas/farmacología
5.
Nat Commun ; 9(1): 4074, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287875

RESUMEN

Persisters are a subpopulation of bacteria that are not killed by antibiotics even though they lack genetic resistance. Here we provide evidence that persisters can manifest as small colony variants (SCVs) in clinical infections. We analyze growth kinetics of Staphylococcus aureus sampled from in vivo conditions and in vitro stress conditions that mimic growth in host compartments. We report that SCVs arise as a result of a long lag time, and that this phenotype emerges de novo during the growth phase in various stress conditions including abscesses and acidic media. We further observe that long lag time correlates with antibiotic usage. These observations suggest that treatment strategies should be carefully tailored to address bacterial persisters in clinics.


Asunto(s)
Absceso/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Animales , Antibacterianos , Proliferación Celular , Humanos , Ratones , Persona de Mediana Edad , Staphylococcus aureus/efectos de los fármacos , Estrés Fisiológico
6.
Appl Environ Microbiol ; 83(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28389547

RESUMEN

Erwinia amylovora is the causative agent of fire blight, a devastating plant disease affecting members of the Rosaceae Alternatives to antibiotics for control of fire blight symptoms and outbreaks are highly desirable, due to increasing drug resistance and tight regulatory restrictions. Moreover, the available diagnostic methods either lack sensitivity, lack speed, or are unable to discriminate between live and dead bacteria. Owing to their extreme biological specificity, bacteriophages are promising alternatives for both aims. In this study, the virulent broad-host-range E. amylovora virus Y2 was engineered to enhance its killing activity and for use as a luciferase reporter phage, respectively. Toward these aims, a depolymerase gene of E. amylovora virus L1 (dpoL1-C) or a bacterial luxAB fusion was introduced into the genome of Y2 by homologous recombination. The genes were placed downstream of the major capsid protein orf68, under the control of the native promoter. The modifications did not affect viability of infectivity of the recombinant viruses. Phage Y2::dpoL1-C demonstrated synergistic activity between the depolymerase degrading the exopolysaccharide capsule and phage infection, which greatly enhanced bacterial killing. It also significantly reduced the ability of E. amylovora to colonize the surface of detached flowers. The reporter phage Y2::luxAB transduced bacterial luciferase into host cells and induced synthesis of large amounts of a LuxAB luciferase fusion. After the addition of aldehyde substrate, bioluminescence could be readily monitored, and this enabled rapid and specific detection of low numbers of viable bacteria, without enrichment, both in vitro and in plant material.IMPORTANCE Fire blight, caused by Erwinia amylovora, is the major threat to global pome fruit production, with high economic losses every year. Bacteriophages represent promising alternatives to not only control the disease, but also for rapid diagnostics. To enhance biocontrol efficacy, we combined the desired properties of two phages, Y2 (broad host range) and L1 (depolymerase for capsule degradation) in a single recombinant phage. This phage showed enhanced biocontrol and could reduce E. amylovora on flowers. Phage Y2 was also genetically engineered into a luciferase reporter phage, which transduces bacterial bioluminescence into infected cells and allows detection of low numbers of viable target bacteria. The combination of speed, sensitivity, and specificity is superior to previously used diagnostic methods. In conclusion, genetic engineering could improve the properties of phage Y2 toward better killing efficacy and sensitive detection of E. amylovora cells.


Asunto(s)
Bacteriófagos/genética , Bacteriófagos/patogenicidad , Erwinia amylovora/virología , Enfermedades de las Plantas/prevención & control , Proteínas Virales/genética , Bacteriófagos/metabolismo , Erwinia amylovora/fisiología , Regulación Viral de la Expresión Génica , Ingeniería Genética , Malus/microbiología , Enfermedades de las Plantas/microbiología , Proteínas Virales/metabolismo , Virulencia
7.
J Infect Dis ; 215(3): 490-491, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974435
8.
Int J Med Microbiol ; 307(1): 11-20, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27955998

RESUMEN

There has been controversy about the intrinsic virulence of methicillin-resistant Staphylococcus aureus (MRSA) as compared to methicillin-susceptible S. aureus (MSSA). To address this discrepancy, the intrinsic virulence of 42 MRSA and 40 MSSA clinical isolates was assessed by testing endothelial cell (EC) damage, a surrogate marker for virulence in blood stream infections. Since these clinical isolates represent a heterogeneous group, well characterized S. aureus laboratory strains with SCCmec loss- and gain-of-function mutations were used in addition. The clinical MRSA isolates carrying typical hospital acquired SCCmec types (I, II or III) induced significantly less damage (47.8%) as compared to isolates with other SCCmec types (62.3%, p=0.03) and MSSA isolates (64.2%, p<0.01). There was a strong inverse correlation between high-level oxacillin resistance and low EC damage induction (R2=0.4464, p<0.001). High-level oxacillin resistant strains (MIC >32µ/ml) grew significantly slower as compared to isolates with low-level resistance (p=0.047). The level of EC damage positively correlated with α- and δ-toxin production (p<0.0001 and p<0.05, respectively) but not with ß-toxin production. Invasive MRSA isolates (n=21, 56.3%) were significantly less cytotoxic as compared to invasive MSSA isolates (n=20, 68.0%, p<0.05). There was no difference between EC damage induced by superficial versus invasive isolates in either MRSA or MSSA strains. Our data suggest that the intrinsic virulence of MRSA is similar or even reduced as compared to MSSA strains but is linked to the level of methicillin resistance.


Asunto(s)
Células Endoteliales/microbiología , Células Endoteliales/fisiología , Interacciones Huésped-Patógeno , Resistencia a la Meticilina , Staphylococcus aureus/fisiología , Antibacterianos/farmacología , Toxinas Bacterianas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromosomas Bacterianos , Eliminación de Gen , Orden Génico , Humanos , Secuencias Repetitivas Esparcidas , Pruebas de Sensibilidad Microbiana , Oxacilina/farmacología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/aislamiento & purificación , Virulencia
9.
J Infect Dis ; 213(2): 305-13, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26188074

RESUMEN

BACKGROUND: Staphylococcus aureus-infected patients treated with antibiotics that are effective in vitro often experience relapse of infection because the bacteria hide in privileged locations. These locations include abscesses and host cells, which contain low-pH compartments and are sites from which nonstable S. aureus small-colony variants (SCVs) are frequently recovered. METHODS: We assessed the effect of low pH on S. aureus colony phenotype and bacterial growth, using in vitro and in vivo models of long-term infection. RESULTS: We showed that low pH induced nonstable SCVs and nonreplicating persisters that are capable of regrowth. Within host cells, S. aureus was located in phagolysosomes, a low-pH compartment. Therapeutic neutralization of phagolysosomal pH with ammonium chloride, bafilomycin A1, or the antimalaria drug chloroquine reduced SCVs in infected host cells. In a systemic mouse infection model, treatment with chloroquine also reduced SCVs. CONCLUSIONS: Our results show that the acidic environment favors formation of nonstable SCVs, which reflect the SCVs found in clinics. They also provide evidence that treatment with alkalinizing agents, together with antibiotics, may provide a novel translational strategy for eradicating persisting intracellular reservoirs of staphylococci. This approach may also be extended to other intracellular bacteria.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Fagosomas/química , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Cloruro de Amonio/farmacología , Animales , Línea Celular Tumoral , Cloroquina/farmacología , Regulación Bacteriana de la Expresión Génica , Variación Genética , Humanos , Concentración de Iones de Hidrógeno , Macrólidos/farmacología , Ratones , Ratones Endogámicos C57BL , Staphylococcus aureus/crecimiento & desarrollo
11.
Infect Control Hosp Epidemiol ; 36(11): 1305-12, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26290400

RESUMEN

OBJECTIVE: In-hospital transmission of methicillin-susceptible Staphylococcus aureus (MSSA) among neonates remains enigmatic. We describe the epidemiology of MSSA colonization and infection in a 30-bed neonatal ward. DESIGN: Multimodal outbreak investigation SETTING: A public 800-bed tertiary care university hospital in Switzerland METHODS: Investigations in 2012-2013, triggered by a MSSA infection cluster, included prospective MSSA infection surveillance, microbiologic screening of neonates and environment, onsite observations, and a prospective cohort study. MSSA isolates were characterized by pulsed-field gel electrophoresis (PFGE) and selected isolates were examined for multilocus sequence type (MLST) and virulence factors. RESULTS: Among 726 in 2012, 30 (4.1%) patients suffered from MSSA infections including 8 (1.1%) with bacteremia. Among 655 admissions in 2013, 13 (2.0%) suffered from MSSA infections including 2 (0.3%) with bacteremia. Among 177 neonates screened for S. aureus carriage, overall 77 (44%) tested positive. A predominant PFGE-1-ST30 strain was identified in 6 of 30 infected neonates (20%) and 30 of 77 colonized neonates (39%). This persistent clone was pvl-negative, tst-positive and belonged to agr group III. We found no environmental point source. MSSA carriage was associated with central vascular catheter use but not with a particular midwife, nurse, physician, or isolette. Observed healthcare worker behavior may have propagated transmission via hands and fomites. Despite multimodal interventions, clonal transmission and colonization continued and another clone, PFGE-6-ST5, became predominant. CONCLUSIONS: Hospital-acquired MSSA clones represent a high proportion of MSSA colonization but not MSSA infections in neonate inpatients. In contrast to persisting MSSA, transmission infection rates decreased concurrently with interventions. It remains to be established whether eradication of hospital-acquired MSSA strains would reduce infection rates further.


Asunto(s)
Bacteriemia/epidemiología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Infecciones Estafilocócicas/epidemiología , Estudios de Cohortes , Electroforesis en Gel de Campo Pulsado , Femenino , Humanos , Lactante , Recién Nacido , Modelos Logísticos , Masculino , Tipificación de Secuencias Multilocus , Análisis Multivariante , Neonatología , Factores de Riesgo , Suiza , Centros de Atención Terciaria
12.
Ann Clin Microbiol Antimicrob ; 14: 14, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25858549

RESUMEN

BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) is a global epidemic threat. The aim of this study was to determine which globally known MRSA lineages are currently present at our tertiary care hospital in Switzerland, a hospital with low MRSA prevalence. In light of the increasing prevalence of multi drug resistance including vancomycin resistance we also assessed antibiotic susceptibilities. METHODS: The 146 MRSA strains collected over two years (March 2012 until February 2014) at the University Hospital Zurich, Switzerland, were analyzed by PFGE analysis of SmaI digests in combination with spa-typing. In addition, representative isolates were analyzed by multi locus sequence typing (MLST). Susceptibilities to eight antibiotics were assessed using the Kirby-Bauer disc diffusion method. RESULTS: Isolates showed resistance to erythromycin (48%), ciprofloxacin (43%), clindamycin (31%), tetracycline (22%), and gentamicin (16%). All isolates were susceptible to vancomycin, 95% were susceptible to sulfamethoxazole/trimethoprim and rifampicin, respectively. PFGE analysis revealed 22 different patterns, with four major patterns that accounted for 53.4% of all MRSA isolates, and seven sporadic patterns. Spa typing revealed 50 different spa types with the predominant types being t008 (14%), t002 (10%), and t127 (9%). 82% of the MRSA isolates could be assigned to six clonal complexes (CCs) namely CC1 (10%), CC5 (23%), CC8 (18%), CC22 (17%), CC30 (11%), and CC45 (3%) based on spa-types, PFGE patterns, and MLST. Two isolates could not be typed by either PFGE analysis or spa-typing and three isolates had spa-types that have not yet been described. CONCLUSIONS: The combination of the two typing methods was more discriminatory as compared to the use of a single method. Several of the lineages that are predominant in Europe are present in our hospital. Resistances to antibiotics have decreased in comparison to a study conducted between 2004 and 2006.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Infecciones Estafilocócicas/microbiología , Técnicas de Tipificación Bacteriana , Farmacorresistencia Bacteriana , Hospitales Universitarios/estadística & datos numéricos , Humanos , Meticilina/farmacología , Staphylococcus aureus Resistente a Meticilina/genética , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/epidemiología , Suiza/epidemiología
13.
Int J Med Microbiol ; 304(8): 1118-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25200859

RESUMEN

USA300 methicillin-resistant Staphylococcus aureus (MRSA) is the most prevalent MRSA in the United States of America (USA) and a global epidemic threat. We investigated the prevalence of USA300 at a tertiary care hospital in Zurich, Switzerland, where all MRSA strains have been collected and PFGE typed since 1992. These strains were retrospectively compared to the PFGE pattern of USA300 strain JE2. Isolates with a respective PFGE pattern were spa-typed and tested for the presence of the arginine catabolic mobile element (ACME) arc gene cluster and Panton-Valentine Leucocidin (PVL) genes. The first MRSA strain with a USA300 PFGE pattern was isolated in 2001 from a patient visiting from the USA. USA300 strains represented between 0% (in 2002) and 9.2% (in 2012) of all MRSA isolates in our hospital. We identified various USA300 subtypes based on either the PFGE pattern, the spa-type or absence of either the PVL genes or ACME arc gene cluster. All the USA300 strains including the variants (n=47) accounted for 5.6% of all MRSA isolates typed between 2001 and 2013 and reached a maximum of 14.5% in 2009. They predominantly caused skin and soft tissue infections (74.4%). In conclusion, even though USA300 has been present in our hospital for over twelve years it has not become the predominant MRSA clone like in the USA. However, in light of the global burden of USA300, care must be taken to further contain the spread of this lineage and of MRSA in general in our hospital.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Tipificación Molecular , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/microbiología , Adulto , Anciano , Electroforesis en Gel de Campo Pulsado , Femenino , Genes Bacterianos , Genotipo , Hospitales , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/clasificación , Staphylococcus aureus Resistente a Meticilina/genética , Persona de Mediana Edad , Prevalencia , Infecciones de los Tejidos Blandos/epidemiología , Infecciones de los Tejidos Blandos/microbiología , Suiza/epidemiología , Adulto Joven
14.
J Proteome Res ; 13(10): 4446-56, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25185497

RESUMEN

Addressing the functionality of predicted genes remains an enormous challenge in the postgenomic era. A prime example of genes lacking functional assignments are the poorly conserved, early expressed genes of lytic bacteriophages, whose products are involved in the subversion of the host metabolism. In this study, we focused on the composition of important macromolecular complexes of Pseudomonas aeruginosa involved in transcription, DNA replication, fatty acid biosynthesis, RNA regulation, energy metabolism, and cell division during infection with members of seven distinct clades of lytic phages. Using affinity purifications of these host protein complexes coupled to mass spectrometric analyses, 37 host complex-associated phage proteins could be identified. Importantly, eight of these show an inhibitory effect on bacterial growth upon episomal expression, suggesting that these phage proteins are potentially involved in hijacking the host complexes. Using complementary protein-protein interaction assays, we further mapped the inhibitory interaction of gp12 of phage 14-1 to the α subunit of the RNA polymerase. Together, our data demonstrate the powerful use of interactomics to unravel the biological role of hypothetical phage proteins, which constitute an enormous untapped source of novel antibacterial proteins. (Data are available via ProteomeXchange with identifier PXD001199.).


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas Virales/metabolismo , Marcadores de Afinidad , Western Blotting , Cromatografía de Afinidad , Unión Proteica , Espectrometría de Masas en Tándem
15.
PLoS One ; 7(8): e43421, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916258

RESUMEN

Extracytoplasmic function (ECF) σ factors control the transcription of genes involved in different cellular functions, such as stress responses, metal homeostasis, virulence-related traits, and cell envelope structure. The genome of Bradyrhizobium japonicum, the nitrogen-fixing soybean endosymbiont, encodes 17 putative ECF σ factors belonging to nine different ECF σ factor families. The genes for two of them, ecfQ (bll1028) and ecfF (blr3038), are highly induced in response to the reactive oxygen species hydrogen peroxide (H(2)O(2)) and singlet oxygen ((1)O(2)). The ecfF gene is followed by the predicted anti-σ factor gene osrA (blr3039). Mutants lacking EcfQ, EcfF plus OsrA, OsrA alone, or both σ factors plus OsrA were phenotypically characterized. While the symbiotic properties of all mutants were indistinguishable from the wild type, they showed increased sensitivity to singlet oxygen under free-living conditions. Possible target genes of EcfQ and EcfF were determined by microarray analyses, and candidate genes were compared with the H(2)O(2)-responsive regulon. These experiments disclosed that the two σ factors control rather small and, for the most part, distinct sets of genes, with about half of the genes representing 13% of the members of H(2)O(2)-responsive regulon. To get more insight into transcriptional regulation of both σ factors, the 5' ends of ecfQ and ecfF mRNA were determined. The presence of conserved sequence motifs in the promoter region of ecfQ and genes encoding EcfQ-like σ factors in related α-proteobacteria suggests regulation via a yet unknown transcription factor. By contrast, we have evidence that ecfF is autoregulated by transcription from an EcfF-dependent consensus promoter, and its product is negatively regulated via protein-protein interaction with OsrA. Conserved cysteine residues 129 and 179 of OsrA are required for normal function of OsrA. Cysteine 179 is essential for release of EcfF from an EcfF-OsrA complex upon H(2)O(2) stress while cysteine 129 is possibly needed for EcfF-OsrA interaction.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bradyrhizobium/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor sigma/metabolismo , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano/genética , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...