Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Bacteriol ; 204(10): e0005422, 2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36106853

RESUMEN

The outer membrane (OM) of Gram-negative bacteria efficiently protects from harmful environmental stresses such as antibiotics, disinfectants, or dryness. The main constituents of the OM are integral OM ß-barrel proteins (OMPs). In Gram-negative bacteria such as Escherichia coli, Yersinia enterocolitica, and Pseudomonas aeruginosa, the insertion of OMPs depends on a sophisticated biogenesis pathway. This comprises the SecYEG translocon, which enables inner membrane (IM) passage; the chaperones SurA, Skp, and DegP, which facilitate the passage of ß-barrel OMPs through the periplasm; and the ß-barrel assembly machinery (BAM), which facilitates insertion into the OM. In E. coli, Y. enterocolitica, and P. aeruginosa, the deletion of SurA is particularly detrimental and leads to a loss of OM integrity, sensitization to antibiotic treatment, and reduced virulence. In search of targets that could be exploited to develop compounds that interfere with OM integrity in Acinetobacter baumannii, we employed the multidrug-resistant strain AB5075 to generate single gene knockout strains lacking individual periplasmic chaperones. In contrast to E. coli, Y. enterocolitica, and P. aeruginosa, AB5075 tolerates the lack of SurA, Skp, or DegP with only weak mutant phenotypes. While the double knockout strains ΔsurAΔskp and ΔsurAΔdegP are conditionally lethal in E. coli, all double deletions were well tolerated by AB5075. Strikingly, even a triple-knockout strain of AB5075, lacking surA, skp, and degP, was viable. IMPORTANCE Acinetobacter baumannii is a major threat to human health due to its ability to persist in the hospital environment, resistance to antibiotic treatment, and ability to deploy multiple and redundant virulence factors. In a rising number of cases, infections with multidrug-resistant A. baumannii end up fatally, because all antibiotic treatment options fail. Thus, novel targets have to be identified and alternative therapeutics have to be developed. The knockout of periplasmic chaperones has previously proven to significantly reduce virulence and even break antibiotic resistance in other Gram-negative pathogens. Our study in A. baumannii demonstrates how variable the importance of the periplasmic chaperones SurA, Skp, and DegP can be and suggests the existence of mechanisms allowing A. baumannii to cope with the lack of the three periplasmic chaperones.


Asunto(s)
Acinetobacter baumannii , Proteínas de la Membrana Bacteriana Externa , Desinfectantes , Humanos , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Infección Hospitalaria/microbiología , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Hospitales , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Periplasma/metabolismo , Pliegue de Proteína , Canales de Translocación SEC/metabolismo , Factores de Virulencia/metabolismo , Yersinia enterocolitica , Pseudomonas aeruginosa , Farmacorresistencia Bacteriana Múltiple
2.
Artículo en Inglés | MEDLINE | ID: mdl-31818817

RESUMEN

With the aim to identify potential new targets to restore antimicrobial susceptibility of multidrug-resistant (MDR) Pseudomonas aeruginosa isolates, we generated a high-density transposon (Tn) insertion mutant library in an MDR P. aeruginosa bloodstream isolate (isolate ID40). The depletion of Tn insertion mutants upon exposure to cefepime or meropenem was measured in order to determine the common resistome for these clinically important antipseudomonal ß-lactam antibiotics. The approach was validated by clean deletions of genes involved in peptidoglycan synthesis/recycling, such as the genes for the lytic transglycosylase MltG, the murein (Mur) endopeptidase MepM1, the MurNAc/GlcNAc kinase AmgK, and the uncharacterized protein YgfB, all of which were identified in our screen as playing a decisive role in survival after treatment with cefepime or meropenem. We found that the antibiotic resistance of P. aeruginosa can be overcome by targeting usually nonessential genes that turn essential in the presence of therapeutic concentrations of antibiotics. For all validated genes, we demonstrated that their deletion leads to the reduction of ampC expression, resulting in a significant decrease in ß-lactamase activity, and consequently, these mutants partly or completely lost resistance against cephalosporins, carbapenems, and acylaminopenicillins. In summary, the determined resistome may comprise promising targets for the development of drugs that may be used to restore sensitivity to existing antibiotics, specifically in MDR strains of P. aeruginosa.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Elementos Transponibles de ADN , Farmacorresistencia Bacteriana Múltiple/genética , Pseudomonas aeruginosa/genética , Resistencia betalactámica/genética , Proteínas Bacterianas/metabolismo , Cefepima/farmacología , Endopeptidasas/deficiencia , Endopeptidasas/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Humanos , Meropenem/farmacología , Pruebas de Sensibilidad Microbiana , Mutagénesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/enzimología , Pseudomonas aeruginosa/aislamiento & purificación , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
3.
Brain Behav Immun ; 87: 329-338, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31904407

RESUMEN

Sleep strongly impacts both humoral and cellular immunity; however, its acute effects on the innate immune defense against pathogens are unclear. Here, we elucidated in mice whether sleep affects the numbers and functions of innate immune cells and their defense against systemic bacterial infection. Sleep significantly increased numbers of classical monocytes in blood and spleen of mice that were allowed to sleep for six hours at the beginning of the normal resting phase compared to mice kept awake for the same time. The sleep-induced effect on classical monocytes was neither caused by alterations in corticosterone nor myelopoiesis, bone marrow egress or death of monocytes and did only partially involve Gαi-protein coupled receptors like chemokine receptor 2 (CCR2), but not the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) or lymphocyte function-associated antigen 1 (LFA-1). Notably, sleep suppressed the expression of the clock gene Arntl in splenic monocytes and the sleep-induced increase in circulating classical monocytes was abrogated in Arntl-deficient animals, indicating that sleep is a prerequisite for clock-gene driven rhythmic trafficking of classical monocytes. Sleep also enhanced the production of reactive oxygen species by monocytes and neutrophils. Moreover, sleep profoundly reduced bacterial load in blood and spleen of mice that were allowed to sleep before systemic bacterial infection and consequently increased survival upon infection. These data provide the first evidence that sleep enhances numbers and function of innate immune cells and therewith strengthens early defense against bacterial pathogens.


Asunto(s)
Infecciones Bacterianas , Monocitos , Animales , Molécula 1 de Adhesión Intercelular , Ratones , Ratones Endogámicos C57BL , Neutrófilos , Sueño
4.
Med Microbiol Immunol ; 209(3): 277-299, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31784893

RESUMEN

The capacity of pathogenic microorganisms to adhere to host cells and avoid clearance by the host immune system is the initial and most decisive step leading to infections. Bacteria have developed different strategies to attach to diverse host surface structures. One important strategy is the adhesion to extracellular matrix (ECM) proteins (e.g., collagen, fibronectin, laminin) that are highly abundant in connective tissue and basement membranes. Gram-negative bacteria express variable outer membrane proteins (adhesins) to attach to the host and to initiate the process of infection. Understanding the underlying molecular mechanisms of bacterial adhesion is a prerequisite for targeting this interaction by "anti-ligands" to prevent colonization or infection of the host. Future development of such "anti-ligands" (specifically interfering with bacteria-host matrix interactions) might result in the development of a new class of anti-infective drugs for the therapy of infections caused by multidrug-resistant Gram-negative bacteria. This review summarizes our current knowledge about the manifold interactions of adhesins expressed by Gram-negative bacteria with ECM proteins and the use of this information for the generation of novel therapeutic antivirulence strategies.


Asunto(s)
Adhesinas Bacterianas/fisiología , Adhesión Bacteriana , Proteínas de la Matriz Extracelular/fisiología , Fibronectinas/fisiología , Bacterias Gramnegativas/fisiología , Interacciones Microbiota-Huesped , Bacterias Gramnegativas/patogenicidad , Humanos
5.
Int J Med Microbiol ; 309(5): 331-337, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31176600

RESUMEN

Bacteria often express numerous virulence factors. These virulence factors make them successful pathogens, by e.g. mediating attachment to host cells and thereby facilitating persistence or invasion, or by contributing to the evasion of the host immune system to allow proliferation and spread within the host and in the environment. The site of first contact of Gram negative bacteria with the host is the bacterial outer membrane (OM). Consisting of an asymmetrical lipid bilayer with phospholipids forming the inner, and lipopolysaccharides forming the outer leaflet, the OM harbors numerous integral membrane proteins that are almost exclusively ß-barrel proteins. One distinct family of OM ß-barrel proteins strongly linked to bacterial virulence are the autotransporter (AT) proteins. During the last years huge progress has been made to better understand the mechanisms underlying the insertion of AT proteins into the OM and also AT function for interaction with the host. This review shortly summarizes our current knowledge about outer membrane protein (OMP) and more specifically AT biogenesis and function. We focused on the AT proteins that we haved studied in most detail: i.e. the Yersinia adhesin A (YadA) and invasin of Yersinia enterocolitica (Ye) as well as its homolog intimin (Int) expressed by enteropathogenic Escherichia coli. In addition, this review provides a short outlook about how we could possibly use this knowledge to fight infection.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Escherichia coli/metabolismo , Sistemas de Secreción Tipo V/metabolismo , Escherichia coli Enteropatógena/metabolismo , Virulencia , Factores de Virulencia/metabolismo , Yersinia enterocolitica/metabolismo
6.
J Biol Chem ; 291(38): 20096-112, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27466361

RESUMEN

Intimin is an essential adhesin of attaching and effacing organisms such as entropathogenic Escherichia coli It is also the prototype of type Ve secretion or inverse autotransport, where the extracellular C-terminal region or passenger is exported with the help of an N-terminal transmembrane ß-barrel domain. We recently reported a stalled secretion intermediate of intimin, where the passenger is located in the periplasm but the ß-barrel is already inserted into the membrane. Stalling of this mutant is due to the insertion of an epitope tag at the very N terminus of the passenger. Here, we examined how this insertion disrupts autotransport and found that it causes misfolding of the N-terminal immunoglobulin (Ig)-like domain D00. We could also stall the secretion by making an internal deletion in D00, and introducing the epitope tag into the second Ig-like domain, D0, also resulted in reduced passenger secretion. In contrast to many classical autotransporters, where a proximal folding core in the passenger is required for secretion, the D00 domain is dispensable, as the passenger of an intimin mutant lacking D00 entirely is efficiently exported. Furthermore, the D00 domain is slightly less stable than the D0 and D1 domains, unfolding at ∼200 piconewtons (pN) compared with ∼250 pN for D0 and D1 domains as measured by atomic force microscopy. Our results support a model where the secretion of the passenger is driven by sequential folding of the extracellular Ig-like domains, leading to vectorial transport of the passenger domain across the outer membrane in an N to C direction.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Biológicos , Pliegue de Proteína , Adhesinas Bacterianas/genética , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Dominios Proteicos
7.
J Biol Chem ; 290(3): 1837-49, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25488660

RESUMEN

Autotransporter proteins comprise a large family of virulence factors that consist of a ß-barrel translocation unit and an extracellular effector or passenger domain. The ß-barrel anchors the protein to the outer membrane of Gram-negative bacteria and facilitates the transport of the passenger domain onto the cell surface. By inserting an epitope tag into the N terminus of the passenger domain of the inverse autotransporter intimin, we generated a mutant defective in autotransport. Using this stalled mutant, we could show that (i) at the time point of stalling, the ß-barrel appears folded; (ii) the stalled autotransporter is associated with BamA and SurA; (iii) the stalled intimin is decorated with large amounts of SurA; (iv) the stalled autotransporter is not degraded by periplasmic proteases; and (v) inverse autotransporter passenger domains are translocated by a hairpin mechanism. Our results suggest a function for the BAM complex not only in insertion and folding of the ß-barrel but also for passenger translocation.


Asunto(s)
Adhesinas Bacterianas/fisiología , Proteínas de Escherichia coli/fisiología , Escherichia coli/metabolismo , Adhesinas Bacterianas/química , Transporte Biológico , Membrana Celular/metabolismo , Clonación Molecular , Reactivos de Enlaces Cruzados/química , Epítopos/química , Proteínas de Escherichia coli/química , Células HeLa , Humanos , Microscopía Fluorescente , Chaperonas Moleculares/química , Mutagénesis Sitio-Dirigida , Mutación , Péptido Hidrolasas/química , Unión Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Propiedades de Superficie
8.
Mol Microbiol ; 95(1): 80-100, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25353290

RESUMEN

Intimin and Invasin are prototypical inverse (Type Ve) autotransporters and important virulence factors of enteropathogenic Escherichia coli and Yersinia spp. respectively. In addition to a C-terminal extracellular domain and a ß-barrel transmembrane domain, both proteins also contain a short N-terminal periplasmic domain that, in Intimin, includes a lysin motif (LysM), which is thought to mediate binding to peptidoglycan. We show that the periplasmic domain of Intimin does bind to peptidoglycan both in vitro and in vivo, but only under acidic conditions. We were able to determine a dissociation constant of 0.8 µM for this interaction, whereas the Invasin periplasmic domain, which lacks a LysM, bound only weakly in vitro and failed to bind peptidoglycan in vivo. We present the solution structure of the Intimin LysM, which has an additional α-helix conserved within inverse autotransporter LysMs but lacking in others. In contrast to previous reports, we demonstrate that the periplasmic domain of Intimin mediates dimerisation. We further show that dimerisation and peptidoglycan binding are general features of LysM-containing inverse autotransporters. Peptidoglycan binding by the periplasmic domain in the infection process may aid in resisting mechanical and chemical stress during transit through the gastrointestinal tract.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Escherichia coli Enteropatógena/metabolismo , Peptidoglicano/metabolismo , Yersinia/metabolismo , Adhesinas Bacterianas/genética , Sitios de Unión , Biología Computacional/métodos , Dimerización , Escherichia coli Enteropatógena/química , Escherichia coli Enteropatógena/genética , Concentración de Iones de Hidrógeno , Modelos Moleculares , Multimerización de Proteína , Estructura Secundaria de Proteína , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Yersinia/química , Yersinia/genética
9.
Int J Med Microbiol ; 306(6): 357-66, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27107739

RESUMEN

Enteropathogenic Yersinia enterocolitica (Ye) enters the host via contaminated food. After colonisation of the small intestine Ye invades the Peyer's patches (PPs) via M cells and disseminates to the mesenteric lymph nodes (MLNs), spleen and liver. Whether Ye uses other invasion routes and which pathogenicity factors are required remains elusive. Oral infection of lymphotoxin-ß-receptor deficient mice lacking PPs and MLNs with Ye revealed similar bacterial load in the spleen 1h post infection as wild-type mice, demonstrating a PP-independent dissemination route for Ye. Immunohistological analysis of the small intestine revealed Ye in close contact with mononuclear phagocytes (MPs), specifically CX3CR1(+) monocyte-derived cells (MCs) as well as CD103(+) dendritic cells (DCs). This finding was confirmed by flow cytometry and imaging flow cytometry analysis of lamina propria (LP) leukocytes showing CD103(+) DCs and MCs with intracellular Ye. Uptake of Ye by LP CD103(+) DCs and MCs was dependent on the pathogenicity factor invasin, whereas the adhesin YadA was dispensable as demonstrated by Ye deletion mutants. Furthermore, Ye were found exclusively associated with CD103(+) DCs in the MLNs from wild-type mice, but not from CCR7(-/-) mice, demonstrating a CCR7 dependent transport of Ye by CD103(+) DCs from LP to the MLNs. In contrast, dissemination of Ye to the spleen was dependent on MCs as significantly less Ye could be recovered from the spleen of CX3CR1(GFP/GFP) mice compared to wild-type mice. Altogether, MCs and CD103(+) DCs contribute to immediate invasion and dissemination of Ye. This together with data from other bacteria suggests MPs as general pathogenic entry site in the intestine.


Asunto(s)
Interacciones Huésped-Patógeno , Intestino Delgado/patología , Fagocitos/microbiología , Yersiniosis/patología , Yersinia enterocolitica/inmunología , Yersinia enterocolitica/fisiología , Animales , Carga Bacteriana , Femenino , Citometría de Flujo , Inmunohistoquímica , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Hígado/microbiología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Bazo/microbiología , Factores de Tiempo , Yersiniosis/inmunología , Yersiniosis/microbiología
10.
Int J Med Microbiol ; 306(2): 77-88, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26718660

RESUMEN

Injection of Yersinia outer proteins (Yops) into host cells by a type III secretion system is an important immune evasion mechanism of Yersinia enterocolitica (Ye). In this process Ye invasin (Inv) binds directly while Yersinia adhesin A (YadA) binds indirectly via extracellular matrix (ECM) proteins to ß1 integrins on host cells. Although leukocytes turned out to be an important target of Yop injection by Ye, it was unclear which Ye adhesins and which leukocyte receptors are required for Yop injection. To explain this, we investigated the role of YadA, Inv and ß1 integrins for Yop injection into leukocytes and their impact on the course of systemic Ye infection in mice. Ex vivo infection experiments revealed that adhesion of Ye via Inv or YadA is sufficient to promote Yop injection into leukocytes as revealed by a ß-lactamase reporter assay. Serum factors inhibit YadA- but not Inv-mediated Yop injection into B and T cells, shifting YadA-mediated Yop injection in the direction of neutrophils and other myeloid cells. Systemic Ye mouse infection experiments demonstrated that YadA is essential for Ye virulence and Yop injection into leukocytes, while Inv is dispensable for virulence and plays only a transient and minor role for Yop injection in the early phase of infection. Ye infection of mice with ß1 integrin-depleted leukocytes demonstrated that ß1 integrins are dispensable for YadA-mediated Yop injection into leukocytes, but contribute to Inv-mediated Yop injection. Despite reduced Yop injection into leukocytes, ß1 integrin-deficient mice exhibited an increased susceptibility for Ye infection, suggesting an important role of ß1 integrins in immune defense against Ye. This study demonstrates that Yop injection into leukocytes by Ye is largely mediated by YadA exploiting, as yet unknown, leukocyte receptors.


Asunto(s)
Adhesinas Bacterianas/fisiología , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Integrina beta1/fisiología , Leucocitos/metabolismo , Yersiniosis/sangre , Yersinia enterocolitica , Adhesinas Bacterianas/genética , Alelos , Animales , Integrina beta1/genética , Ratones , Ratones Endogámicos C57BL , Plásmidos
11.
Cell Microbiol ; 17(8): 1179-204, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25678064

RESUMEN

The current paradigm suggests that Yersinia enterocolitica (Ye) adheres to host cells via the outer membrane proteins Yersinia adhesin A (YadA) or invasin (Inv) to facilitate injection of Yops by the type III secretion system. In this process Inv binds directly to ß1 integrins of host cells while YadA may bind indirectly via extracellular matrix proteins to ß1 integrins. Here we challenged this paradigm and investigated the requirements for Yop injection. We demonstrate that Inv- but not YadA-mediated adhesion depends on ß1 integrin binding and activation, and that tight adhesion is a prerequisite for Yop injection. By means of novel transgenic cell lines, shRNA approaches and RGD peptides, we found that YadA, in contrast to Inv, may use a broad host cell receptor repertoire for host cell adhesion. In the absence of ß1 integrins, YadA mediates Yop injection by interaction with αV integrins in cooperation with yet unknown cofactors expressed by epithelial cells, but not fibroblasts. Electron microscopic and flow chamber studies revealed that a defined intimate contact area between Ye and host cells resulting in adhesion forces resisting shear stress is required for Yop injection. Thus, the indirect binding of YadA to a broad extracellular matrix (ECM) binding host cell receptor repertoire of different cell types makes YadA a versatile tool to ensure Yop injection. In conclusion, given the differential expression of the outer membrane proteins Inv and YadA in the course of Ye infection and differential expression of integrins by various host cell populations, the data demonstrate that Ye is flexibly armed to accomplish Yop injection in different host cell types, a central event in its immune evasion strategy.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Toxinas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Yersinia enterocolitica/fisiología , Células Epiteliales/microbiología , Fibroblastos/metabolismo , Citometría de Flujo , Integrina alfaV/metabolismo , Integrina beta1/metabolismo , Microscopía Electrónica , Unión Proteica , Transporte de Proteínas
12.
J Biol Chem ; 289(43): 29457-70, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25190806

RESUMEN

Yersinia adhesin A (YadA) belongs to a class of bacterial adhesins that form trimeric structures. Their mature form contains a passenger domain and a C-terminal ß-domain that anchors the protein in the outer membrane (OM). Little is known about how precursors of such proteins cross the periplasm and assemble into the OM. In the present study we took advantage of the evolutionary conservation in the biogenesis of ß-barrel proteins between bacteria and mitochondria. We previously observed that upon expression in yeast cells, bacterial ß-barrel proteins including the transmembrane domain of YadA assemble into the mitochondrial OM. In the current study we found that when expressed in yeast cells both the monomeric and trimeric forms of full-length YadA were detected in mitochondria but only the trimeric species was fully integrated into the OM. The oligomeric form was exposed on the surface of the organelle in its native conformation and maintained its capacity to adhere to host cells. The co-expression of YadA with a mitochondria-targeted form of the bacterial periplasmic chaperone Skp, but not with SurA or SecB, resulted in enhanced levels of both forms of YadA. Taken together, these results indicate that the proper assembly of trimeric autotransporter can occur also in a system lacking the lipoproteins of the BAM machinery and is specifically enhanced by the chaperone Skp.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Secuencia Conservada , Evolución Molecular , Mitocondrias/metabolismo , Multimerización de Proteína , Adhesinas Bacterianas/química , Células HeLa , Humanos , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteolisis , Saccharomyces cerevisiae/metabolismo
13.
J Biol Chem ; 289(11): 7388-98, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24369174

RESUMEN

Trimeric autotransporter adhesins (TAAs) are important virulence factors of many Gram-negative bacterial pathogens. TAAs form fibrous, adhesive structures on the bacterial cell surface. Their N-terminal extracellular domains are exported through a C-terminal membrane pore; the insertion of the pore domain into the bacterial outer membrane follows the rules of ß-barrel transmembrane protein biogenesis and is dependent on the essential Bam complex. We have recently described the full fiber structure of SadA, a TAA of unknown function in Salmonella and other enterobacteria. In this work, we describe the structure and function of SadB, a small inner membrane lipoprotein. The sadB gene is located in an operon with sadA; orthologous operons are only found in enterobacteria, whereas other TAAs are not typically associated with lipoproteins. Strikingly, SadB is also a trimer, and its co-expression with SadA has a direct influence on SadA structural integrity. This is the first report of a specific export factor of a TAA, suggesting that at least in some cases TAA autotransport is assisted by additional periplasmic proteins.


Asunto(s)
Enterobacteriaceae/metabolismo , Lipoproteínas/metabolismo , Salmonella/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Transporte Biológico , Separación Celular , Clonación Molecular , Cartilla de ADN , Citometría de Flujo , Lipoproteínas/genética , Modelos Moleculares , Biblioteca de Péptidos , Periplasma/metabolismo , Plásmidos/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Propiedades de Superficie
14.
Antimicrob Agents Chemother ; 59(12): 7335-45, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26369961

RESUMEN

The human gut forms a dynamic reservoir of antibiotic resistance genes (ARGs). Treatment with antimicrobial agents has a significant impact on the intestinal resistome and leads to enhanced horizontal transfer and selection of resistance. We have monitored the development of intestinal ARGs over a 6-day course of ciprofloxacin (Cp) treatment in two healthy individuals by using sequenced-based metagenomics and different ARG quantification methods. Fixed- and random-effect models were applied to determine the change in ARG abundance per defined daily dose of Cp as an expression of the respective selection pressure. Among various shifts in the composition of the intestinal resistome, we found in one individual a strong positive selection for class D beta-lactamases which were partly located on a mobile genetic element. Furthermore, a trend to a negative selection has been observed with class A beta-lactamases (-2.66 hits per million sample reads/defined daily dose; P = 0.06). By 4 weeks after the end of treatment, the composition of ARGs returned toward their initial state but to a different degree in both subjects. We present here a novel analysis algorithm for the determination of antibiotic selection pressure which can be applied in clinical settings to compare therapeutic regimens regarding their effect on the intestinal resistome. This information is of critical importance for clinicians to choose antimicrobial agents with a low selective force on their patients' intestinal ARGs, likely resulting in a diminished spread of resistance and a reduced burden of hospital-acquired infections with multidrug-resistant pathogens.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Microbioma Gastrointestinal/efectos de los fármacos , Metagenómica/métodos , Adulto , Algoritmos , Biodiversidad , Ciprofloxacina/farmacología , Microbioma Gastrointestinal/genética , Humanos , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Selección Genética/efectos de los fármacos , beta-Lactamasas/genética
15.
Int J Med Microbiol ; 305(2): 276-82, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25596886

RESUMEN

Intimin and invasin are adhesins and central virulence factors of attaching and effacing bacteria, such as enterohaemorrhagic Escherichia coli, and enteropathogenic Yersiniae, respectively. These proteins are prototypes of a large family of adhesins distributed widely in Gram-negative bacteria. It is now evident that this protein family represents a previously unrecognized autotransporter secretion system, termed type Ve secretion. In contrast to classical autotransport, where the transmembrane ß-barrel domain or translocation unit is C-terminal to the extracellular region or passenger domain, type Ve-secreted proteins have an inverted topology with the passenger domain C-terminal to the translocation unit; hence the term inverse autotransporter. This minireview covers the recent advances in elucidating the structure and biogenesis of inverse autotransporters.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Bacterias Gramnegativas/química , Bacterias Gramnegativas/metabolismo , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Factores de Virulencia/química , Factores de Virulencia/metabolismo
16.
Int J Med Microbiol ; 305(2): 252-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25604505

RESUMEN

The trimeric autotransporter adhesin Yersinia adhesin A is the prototype of the type Vc secretion systems. It is expressed by enteropathogenic Yersinia enterocolitica and Yersinia pseudotuberculosis strains, but not by Yersinia pestis. A characteristic trait of YadA is its modular composition and trimeric nature. YadA consists of an N-terminal passenger domain which is exposed on the bacterial cell surface. The translocation of this passenger onto the surface is facilitated by a C-terminal ß-barrel domain which concomitantly anchors YadA into the outer membrane with three YadA monomers contributing to the formation of a single ß-barrel. In Y. enterocolitica, but not Y. pseudotuberculosis, YadA is a decisive virulence factor and its deletion renders the bacteria virtually avirulent in mouse models of infection. This striking importance of YadA in infection may derive from its manifold functions in host cell interaction. Presumably the most important function of YadA is that it mediates adhesion to extracellular matrix components of eukaryotic host cells. Only tight adhesion allows for the injection of "anti-host" effector proteins via a type III secretion system into the host cell cytosol. These effector proteins enable Yersinia to subvert the host immune system in order to replicate and establish infection. YadA is also essential for the survival of Y. enterocolitica upon contact with serum, an important immune-evasion mechanism called serum resistance. To this end, YadA interacts with several components of the host complement system, the first line of immune defense. This review will summarize recent findings about the structure and biogenesis of YadA and its interactions with the host complement system.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Sistemas de Secreción Bacterianos , Factores de Virulencia/metabolismo , Yersinia enterocolitica/fisiología , Yersinia pseudotuberculosis/fisiología , Animales , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Virulencia , Yersinia enterocolitica/crecimiento & desarrollo , Yersinia enterocolitica/metabolismo , Yersinia pseudotuberculosis/metabolismo
17.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 6): 1779-89, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24914988

RESUMEN

Outer membrane protein (OMP) biogenesis is an essential process for maintaining the bacterial cell envelope and involves the ß-barrel assembly machinery (BAM) for OMP recognition, folding and assembly. In Escherichia coli this function is orchestrated by five proteins: the integral outer membrane protein BamA of the Omp85 superfamily and four associated lipoproteins. To unravel the mechanism underlying OMP folding and insertion, the structure of the E. coli BamA ß-barrel and P5 domain was determined at 3 Šresolution. These data add information beyond that provided in the recently published crystal structures of BamA from Haemophilus ducreyi and Neisseria gonorrhoeae and are a valuable basis for the interpretation of pertinent functional studies. In an `open' conformation, E. coli BamA displays a significant degree of flexibility between P5 and the barrel domain, which is indicative of a multi-state function in substrate transfer. E. coli BamA is characterized by a discontinuous ß-barrel with impaired ß1-ß16 strand interactions denoted by only two connecting hydrogen bonds and a disordered C-terminus. The 16-stranded barrel surrounds a large cavity which implies a function in OMP substrate binding and partial folding. These findings strongly support a mechanism of OMP biogenesis in which substrates are partially folded inside the barrel cavity and are subsequently released laterally into the lipid bilayer.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido
18.
J Immunol ; 189(10): 4900-8, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23071281

RESUMEN

Yersinia adhesin A (YadA) is a major virulence factor of Yersinia enterocolitica. YadA mediates host cell binding and autoaggregation and protects the pathogen from killing by the complement system. Previous studies demonstrated that YadA is the most important single factor mediating serum resistance of Y. enterocolitica, presumably by binding C4b binding protein (C4BP) and factor H, which are both complement inhibitors. Factor H acts as a cofactor for factor I-mediated cleavage of C3b into the inactive form iC3b and thus prevents formation of inflammatory effector compounds and the terminal complement complex. In this study, we challenged the current direct binding model of factor H to YadA and show that Y. enterocolitica YadA recruits C3b and iC3b directly, without the need of an active complement cascade or additional serum factors. Enhanced binding of C3b does not decrease survival of YadA-expressing Yersiniae because C3b becomes readily inactivated by factor H and factor I. Binding of factor H to YadA is greatly reduced in the absence of C3. Experiments using Yersinia lacking YadA or expressing YadA with reduced trimeric stability clearly demonstrate that both the presence and full trimeric stability of YadA are essential for complement resistance. A novel mechanism of factor H binding is presented in which YadA exploits recruitment of C3b or iC3b to attract large amounts of factor H. As a consequence, formation of the terminal complement complex is limited and bacterial survival is enhanced. These findings add a new aspect of how Y. enterocolitica effectively evades the host complement system.


Asunto(s)
Adhesinas Bacterianas/inmunología , Activación de Complemento , Complemento C3/inmunología , Evasión Inmune , Yersinia enterocolitica/inmunología , Adhesinas Bacterianas/genética , Complemento C3/antagonistas & inhibidores , Complemento C3/genética , Proteína de Unión al Complemento C4b , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/inmunología , Humanos , Unión Proteica , Yersinia enterocolitica/genética
19.
Commun Biol ; 6(1): 254, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36894667

RESUMEN

YgfB-mediated ß-lactam resistance was recently identified in multi drug resistant Pseudomonas aeruginosa. We show that YgfB upregulates expression of the ß-lactamase AmpC by repressing the function of the regulator of the programmed cell death pathway AlpA. In response to DNA damage, the antiterminator AlpA induces expression of the alpBCDE autolysis genes and of the peptidoglycan amidase AmpDh3. YgfB interacts with AlpA and represses the ampDh3 expression. Thus, YgfB indirectly prevents AmpDh3 from reducing the levels of cell wall-derived 1,6-anhydro-N-acetylmuramyl-peptides, required to induce the transcriptional activator AmpR in promoting the ampC expression and ß-lactam resistance. Ciprofloxacin-mediated DNA damage induces AlpA-dependent production of AmpDh3 as previously shown, which should reduce ß-lactam resistance. YgfB, however, counteracts the ß-lactam enhancing activity of ciprofloxacin by repressing ampDh3 expression and lowering the benefits of this drug combination. Altogether, YgfB represents an additional player in the complex regulatory network of AmpC regulation.


Asunto(s)
Pseudomonas aeruginosa , Resistencia betalactámica , Pseudomonas aeruginosa/genética , Resistencia betalactámica/genética , Ciprofloxacina/farmacología , beta-Lactamas/farmacología
20.
Biology (Basel) ; 11(2)2022 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-35205164

RESUMEN

The complex interplay of a pathogen with its virulence and fitness factors, the host's immune response, and the endogenous microbiome determine the course and outcome of gastrointestinal infection. The expansion of a pathogen within the gastrointestinal tract implies an increased risk of developing severe systemic infections, especially in dysbiotic or immunocompromised individuals. We developed a mechanistic computational model that calculates and simulates such scenarios, based on an ordinary differential equation system, to explain the bacterial population dynamics during gastrointestinal infection. For implementing the model and estimating its parameters, oral mouse infection experiments with the enteropathogen, Yersinia enterocolitica (Ye), were carried out. Our model accounts for specific pathogen characteristics and is intended to reflect scenarios where colonization resistance, mediated by the endogenous microbiome, is lacking, or where the immune response is partially impaired. Fitting our data from experimental mouse infections, we can justify our model setup and deduce cues for further model improvement. The model is freely available, in SBML format, from the BioModels Database under the accession number MODEL2002070001.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA