Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.047
Filtrar
1.
Gut Microbes ; 14(1): 2004798, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35086419

RESUMEN

Candida albicans (C. albicans) is an opportunistic pathogen causing infections ranging from superficial to life-threatening disseminated infections. In a susceptible host, C. albicans is able to translocate through the gut barrier, promoting its dissemination into deeper organs. C. albicans hyphae can invade human epithelial cells by two well-documented mechanisms: epithelial-driven endocytosis and C. albicans-driven active penetration. One mechanism by which host cells protect themselves against intracellular C. albicans is termed autophagy. The protective role of autophagy during C. albicans infection has been investigated in myeloid cells; however, far less is known regarding the role of this process during the infection of epithelial cells. In the present study, we investigated the role of autophagy-related proteins during the infection of epithelial cells, including intestinal epithelial cells and gut explants, by C. albicans. Using cell imaging, we show that key molecular players of the autophagy machinery (LC3-II, PI3P, ATG16L1, and WIPI2) were recruited at Candida invasion sites. We deepened these observations by electron microscopy analyses that reveal the presence of autophagosomes in the vicinity of invading hyphae. Importantly, these events occur during active penetration of C. albicans into host cells and are associated with plasma membrane damage. In this context, we show that the autophagy-related key proteins ATG5 and ATG16L1 contribute to plasma membrane repair mediated by lysosomal exocytosis and participate in protecting epithelial cells against C. albicans-induced cell death. Our findings provide a novel mechanism by which epithelial cells, forming the first line of defense against C. albicans in the gut, can react to limit C. albicans invasion.


Asunto(s)
Autofagia , Candida albicans/fisiología , Candidiasis/microbiología , Membrana Celular/microbiología , Células Epiteliales/microbiología , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Candida albicans/genética , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/fisiopatología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo
2.
PLoS Genet ; 17(12): e1009586, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34941903

RESUMEN

The cell envelope is essential for viability in all domains of life. It retains enzymes and substrates within a confined space while providing a protective barrier to the external environment. Destabilising the envelope of bacterial pathogens is a common strategy employed by antimicrobial treatment. However, even in one of the best studied organisms, Escherichia coli, there remain gaps in our understanding of how the synthesis of the successive layers of the cell envelope are coordinated during growth and cell division. Here, we used a whole-genome phenotypic screen to identify mutants with a defective cell envelope. We report that loss of yhcB, a conserved gene of unknown function, results in loss of envelope stability, increased cell permeability and dysregulated control of cell size. Using whole genome transposon mutagenesis strategies, we report the comprehensive genetic interaction network of yhcB, revealing all genes with a synthetic negative and a synthetic positive relationship. These genes include those previously reported to have a role in cell envelope biogenesis. Surprisingly, we identified genes previously annotated as essential that became non-essential in a ΔyhcB background. Subsequent analyses suggest that YhcB functions at the junction of several envelope biosynthetic pathways coordinating the spatiotemporal growth of the cell, highlighting YhcB as an as yet unexplored antimicrobial target.


Asunto(s)
Pared Celular/genética , Proteínas de Escherichia coli/genética , Lipopolisacáridos/genética , Oxidorreductasas/genética , Peptidoglicano/genética , División Celular/genética , Membrana Celular/genética , Membrana Celular/microbiología , Pared Celular/microbiología , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Lipopolisacáridos/biosíntesis , Mutagénesis , Fosfolípidos/biosíntesis , Fosfolípidos/genética
3.
mBio ; 12(6): e0293921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34781738

RESUMEN

Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.


Asunto(s)
Actinas/metabolismo , Membrana Celular/microbiología , Forminas/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Caveolina 1/genética , Caveolina 1/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Filaminas/genética , Filaminas/metabolismo , Forminas/genética , Células HeLa , Humanos , Listeria monocytogenes/genética , Listeriosis/genética , Listeriosis/microbiología
4.
mBio ; 12(6): e0302121, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34809452

RESUMEN

Shigella spp. are human bacterial pathogens that cause bacillary dysentery. Virulence depends on a type 3 secretion system (T3SS), a highly conserved structure present in multiple important human and plant pathogens. Upon host cell contact, the T3SS translocon is delivered to the host membrane, facilitates bacterial docking to the membrane, and enables delivery of effector proteins into the host cytosol. The Shigella translocon is composed of two proteins, IpaB and IpaC, which together form this multimeric structure within host plasma membranes. Upon interaction of IpaC with host intermediate filaments, the translocon undergoes a conformational change that allows for bacterial docking onto the translocon and, together with host actin polymerization, enables subsequent effector translocation through the translocon pore. To generate additional insights into the translocon, we mapped the topology of IpaB in plasma membrane-embedded pores using cysteine substitution mutagenesis coupled with site-directed labeling and proximity-enabled cross-linking by membrane-permeant sulfhydryl reactants. We demonstrate that IpaB function is dependent on posttranslational modification by a plasmid-encoded acyl carrier protein. We show that the first transmembrane domain of IpaB lines the interior of the translocon pore channel such that the IpaB portion of the channel forms a funnel-like shape leading into the host cytosol. In addition, we identify regions of IpaB within its cytosolic domain that protrude into and are closely associated with the pore channel. Taken together, these results provide a framework for how IpaB is arranged within translocons natively delivered by Shigella during infection. IMPORTANCE Type 3 secretion systems are nanomachines employed by many bacteria, including Shigella, which deliver into human cells bacterial virulence proteins that alter cellular function in ways that promote infection. Delivery of Shigella virulence proteins occurs through a pore formed in human cell membranes by the IpaB and IpaC proteins. Here, we define how IpaB contributes to the formation of pores natively delivered into human cell membranes by Shigella flexneri. We show that a specific domain of IpaB (transmembrane domain 1) lines much of the pore channel and that portions of IpaB that lie in the inside of the human cell loop back into and/or are closely associated with the pore channel. These findings provide new insights into the organization and function of the pore in serving as the conduit for delivery of virulence proteins into human cells during Shigella infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/microbiología , Disentería Bacilar/microbiología , Shigella flexneri/metabolismo , Transferasas/metabolismo , Sistemas de Secreción Tipo III/química , Sistemas de Secreción Tipo III/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Membrana Celular/química , Humanos , Dominios Proteicos , Shigella flexneri/química , Shigella flexneri/genética , Transferasas/genética , Sistemas de Secreción Tipo III/genética
5.
mBio ; 12(4): e0139221, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34340544

RESUMEN

Pathogen attachment to host tissue is critical in the progress of many infections. Bacteria use adhesion in vivo to stabilize colonization and subsequently regulate the deployment of contact-dependent virulence traits. To specifically target host cells, they decorate themselves with adhesins, proteins that bind to mammalian cell surface receptors. One common assumption is that adhesin-receptor interactions entirely govern bacterial attachment. However, how adhesins engage with their receptors in an in vivo-like context remains unclear, in particular under the influence of a heterogeneous mechanical microenvironment. We here investigate the biophysical processes governing bacterial adhesion to host cells using a tunable adhesin-receptor system. By dynamically visualizing attachment, we found that bacterial adhesion to host cell surface, unlike adhesion to inert surfaces, involves two consecutive steps. Bacteria initially attach to their host without engaging adhesins. This step lasts about 1 min, during which bacteria can easily detach. We found that at this stage, the glycocalyx, a layer of glycosylated proteins and lipids, shields the host cell by keeping adhesins away from their receptor ligand. In a second step, adhesins engage with their target receptors to strengthen attachment for minutes to hours. The active properties of the membrane, endowed by the actin cytoskeleton, strengthen specific adhesion. Altogether, our results demonstrate that adhesin-ligand binding is not the sole regulator of bacterial adhesion. In fact, the host cell's surface mechanical microenvironment mediates the physical interactions between host and bacteria, thereby playing an essential role in the onset of infection. IMPORTANCE Microbial adhesion to host cells is the initial step toward many infections. Despite playing a pivotal role in the onset of disease, we still know little about how bacteria attach in an in vivo-like context. By employing a biophysical approach where we investigated host-microbe physical interactions at the single-cell level, we unexpectedly discovered that bacteria attach to mammalian cell membranes in two successive steps. We found that mechanical factors of the cell microenvironment regulate each of these steps, and even dominate biochemical factors, thereby challenging preconceptions on how pathogens interact with their hosts.


Asunto(s)
Bacterias/metabolismo , Adhesión Bacteriana , Membrana Celular/microbiología , Interacciones Huésped-Patógeno/fisiología , Receptores de Superficie Celular/metabolismo , Adhesinas Bacterianas/metabolismo , Bacterias/clasificación , Bacterias/patogenicidad , Fenómenos Fisiológicos Bacterianos , Membrana Celular/fisiología , Microambiente Celular , Escherichia coli/metabolismo , Células HeLa , Humanos , Receptores Inmunológicos/metabolismo , Virulencia
6.
Front Immunol ; 12: 670205, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248949

RESUMEN

Mast cells are potent immune sensors of the tissue microenvironment. Within seconds of activation, they release various preformed biologically active products and initiate the process of de novo synthesis of cytokines, chemokines, and other inflammatory mediators. This process is regulated at multiple levels. Besides the extensively studied IgE and IgG receptors, toll-like receptors, MRGPR, and other protein receptor signaling pathways, there is a critical activation pathway based on cholesterol-dependent, pore-forming cytolytic exotoxins produced by Gram-positive bacterial pathogens. This pathway is initiated by binding the exotoxins to the cholesterol-rich membrane, followed by their dimerization, multimerization, pre-pore formation, and pore formation. At low sublytic concentrations, the exotoxins induce mast cell activation, including degranulation, intracellular calcium concentration changes, and transcriptional activation, resulting in production of cytokines and other inflammatory mediators. Higher toxin concentrations lead to cell death. Similar activation events are observed when mast cells are exposed to sublytic concentrations of saponins or some other compounds interfering with the membrane integrity. We review the molecular mechanisms of mast cell activation by pore-forming bacterial exotoxins, and other compounds inducing cholesterol-dependent plasma membrane perturbations. We discuss the importance of these signaling pathways in innate and acquired immunity.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Citotoxinas/metabolismo , Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/metabolismo , Mastocitos/metabolismo , Animales , Señalización del Calcio , Muerte Celular , Degranulación de la Célula , Membrana Celular/inmunología , Membrana Celular/microbiología , Membrana Celular/patología , Microambiente Celular , Citocinas/metabolismo , Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/patología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Mastocitos/inmunología , Mastocitos/microbiología , Mastocitos/patología
7.
Toxins (Basel) ; 13(4)2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918753

RESUMEN

Clostridium botulinum C2 toxin is a clostridial binary toxin consisting of actin ADP-ribosyltransferase (C2I) and C2II binding components. Activated C2II (C2IIa) binds to cellular receptors and forms oligomer in membrane rafts. C2IIa oligomer assembles with C2I and contributes to the transport of C2I into the cytoplasm of host cells. C2IIa induces Ca2+-induced lysosomal exocytosis, extracellular release of the acid sphingomyelinase (ASMase), and membrane invagination and endocytosis through generating ceramides in the membrane by ASMase. Here, we reveal that C2 toxin requires the lysosomal enzyme cathepsin B (CTSB) during endocytosis. Lysosomes are a rich source of proteases, containing cysteine protease CTSB and cathepsin L (CTSL), and aspartyl protease cathepsin D (CTSD). Cysteine protease inhibitor E64 blocked C2 toxin-induced cell rounding, but aspartyl protease inhibitor pepstatin-A did not. E64 inhibited the C2IIa-promoted extracellular ASMase activity, indicating that the protease contributes to the activation of ASMase. C2IIa induced the extracellular release of CTSB and CTSL, but not CTSD. CTSB knockdown by siRNA suppressed C2 toxin-caused cytotoxicity, but not siCTSL. These findings demonstrate that CTSB is important for effective cellular entry of C2 toxin into cells through increasing ASMase activity.


Asunto(s)
Toxinas Botulínicas/metabolismo , Catepsina B/metabolismo , Membrana Celular/enzimología , Clostridium botulinum/metabolismo , Endocitosis , Lisosomas/enzimología , Animales , Catepsina B/genética , Membrana Celular/microbiología , Clostridium botulinum/patogenicidad , Perros , Exocitosis , Interacciones Huésped-Patógeno , Lisosomas/genética , Lisosomas/microbiología , Células de Riñón Canino Madin Darby , Esfingomielina Fosfodiesterasa/metabolismo
8.
PLoS One ; 16(3): e0246651, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33651833

RESUMEN

Engulfment requires the coordinated, targeted synthesis and degradation of peptidoglycan at the leading edge of the engulfing membrane to allow the mother cell to completely engulf the forespore. Proteins such as the DMP and Q:AH complexes in Bacillus subtilis are essential for engulfment, as are a set of accessory proteins including GerM and SpoIIB, among others. Experimental and bioinformatic studies of these proteins in bacteria distinct from Bacillus subtilis indicate that fundamental differences exist regarding the organization and mechanisms used to successfully perform engulfment. As a consequence, the distribution and prevalence of the proteins involved in engulfment and other proteins that participate in different sporulation stages have been studied using bioinformatic approaches. These works are based on the prediction of orthologs in the genomes of representative Firmicutes and have been helpful in tracing hypotheses about the origin and evolution of sporulation genes, some of which have been postulated as sporulation signatures. To date, an extensive study of these signatures outside of the representative Firmicutes is not available. Here, we asked whether phyletic profiles of proteins involved in engulfment can be used as signatures able to describe the sporulation phenotype. We tested this hypothesis in a set of 954 Firmicutes, finding preserved phyletic profiles defining signatures at the genus level. Finally, a phylogenetic reconstruction based on non-redundant phyletic profiles at the family level shows the non-monophyletic origin of these proteins due to gain/loss events along the phylum Firmicutes.


Asunto(s)
Membrana Celular/metabolismo , Membrana Celular/microbiología , Genómica , Peptidoglicano/metabolismo , Bacillus subtilis/genética , Pared Celular/metabolismo , Esporas Bacterianas/genética
9.
Sci Rep ; 11(1): 5624, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707642

RESUMEN

Burkholderia cenocepacia is known for its capacity of adherence and interaction with the host, causing severe opportunistic lung infections in cystic fibrosis patients. In this work we produced Giant Plasma Membrane Vesicles (GPMVs) from a bronchial epithelial cell line and validated their use as a cell-like alternative to investigate the steps involved in the adhesion process of B. cenocepacia. RNA-sequencing was performed and the analysis of the B. cenocepacia K56-2 transcriptome after the first contacts with the surface of host cells allowed the recognition of genes implicated in bacterial adaptation and virulence-associated functions. The sensing of host membranes led to a transcriptional shift that caused a cascade of metabolic and physiological adaptations to the host specific environment. Many of the differentially expressed genes encode proteins related with central metabolic pathways, transport systems, cellular processes, and virulence traits. The understanding of the changes in gene expression that occur in the early steps of infection can uncover new proteins implicated in B. cenocepacia-host cell adhesion, against which new blocking agents could be designed to control the progression of the infectious process.


Asunto(s)
Bronquios/patología , Burkholderia cenocepacia/genética , Membrana Celular/microbiología , Células Epiteliales/microbiología , Transcriptoma/genética , Burkholderia cenocepacia/patogenicidad , Adhesión Celular , Línea Celular , Membrana Celular/ultraestructura , Análisis por Conglomerados , Células Epiteliales/ultraestructura , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Humanos , Redes y Vías Metabólicas/genética , Modelos Biológicos
10.
Nat Commun ; 12(1): 1907, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772012

RESUMEN

Prokaryotic cell transcriptomics has been limited to mixed or sub-population dynamics and individual cells within heterogeneous populations, which has hampered further understanding of spatiotemporal and stage-specific processes of prokaryotic cells within complex environments. Here we develop a 'TRANSITomic' approach to profile transcriptomes of single Burkholderia pseudomallei cells as they transit through host cell infection at defined stages, yielding pathophysiological insights. We find that B. pseudomallei transits through host cells during infection in three observable stages: vacuole entry; cytoplasmic escape and replication; and membrane protrusion, promoting cell-to-cell spread. The B. pseudomallei 'TRANSITome' reveals dynamic gene-expression flux during transit in host cells and identifies genes that are required for pathogenesis. We find several hypothetical proteins and assign them to virulence mechanisms, including attachment, cytoskeletal modulation, and autophagy evasion. The B. pseudomallei 'TRANSITome' provides prokaryotic single-cell transcriptomics information enabling high-resolution understanding of host-pathogen interactions.


Asunto(s)
Proteínas Bacterianas/genética , Burkholderia pseudomallei/genética , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Genes Bacterianos/genética , Factores de Virulencia/genética , Animales , Burkholderia pseudomallei/citología , Burkholderia pseudomallei/patogenicidad , Línea Celular Tumoral , Membrana Celular/microbiología , Citoplasma/microbiología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Melioidosis/microbiología , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Análisis de la Célula Individual/métodos , Vacuolas/microbiología , Virulencia/genética
11.
J Mol Biol ; 433(7): 166836, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33539891

RESUMEN

The anaerobic Gram-negative bacterium Porphyromonas gingivalis is considered the keystone of periodontitis diseases, a set of inflammatory conditions that affects the tissues surrounding the teeth. In the recent years, the major virulence factors exploited by P. gingivalis have been identified and characterized, including a cocktail of toxins, mainly proteases called gingipains, which promote gingival tissue invasion. These effectors use the Sec pathway to cross the inner membrane and are then recruited and transported across the outer membrane by the type IX secretion system (T9SS). In P. gingivalis, most secreted effectors are attached to anionic lipopolysaccharides (A-LPS), and hence form a virulence coat at the cell surface. P. gingivalis produces additional virulence factors to evade host immune responses, such as capsular polysaccharide, fimbriae and outer membrane vesicles. In addition to periodontitis, it is proposed that this broad repertoire of virulence factors enable P. gingivalis to be involved in diverse human diseases such as rheumatoid arthritis, and neurodegenerative, Alzheimer, and cardiovascular disorders. Here, we review the major virulence determinants of P. gingivalis and discuss future directions to better understand their mechanisms of action.


Asunto(s)
Sistemas de Secreción Bacterianos/genética , Infecciones por Bacteroidaceae/genética , Cisteína-Endopeptidasas Gingipaínas/genética , Porphyromonas gingivalis/genética , Adhesinas Bacterianas/genética , Infecciones por Bacteroidaceae/microbiología , Membrana Celular/microbiología , Humanos , Lipopolisacáridos/genética , Periodontitis/genética , Periodontitis/microbiología , Porphyromonas gingivalis/patogenicidad , Factores de Virulencia/genética
12.
Toxins (Basel) ; 13(2)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572271

RESUMEN

With the rapid growth of antibiotic-resistant bacteria, it is urgent to develop alternative therapeutic strategies. Pore-forming toxins (PFTs) belong to the largest family of virulence factors of many pathogenic bacteria and constitute the most characterized classes of pore-forming proteins (PFPs). Recent studies revealed the structural basis of several PFTs, both as soluble monomers, and transmembrane oligomers. Upon interacting with host cells, the soluble monomer of bacterial PFTs assembles into transmembrane oligomeric complexes that insert into membranes and affect target cell-membrane permeability, leading to diverse cellular responses and outcomes. Herein we have reviewed the structural basis of pore formation and interaction of PFTs with the host cell membrane, which could add valuable contributions in comprehensive understanding of PFTs and searching for novel therapeutic strategies targeting PFTs and interaction with host receptors in the fight of bacterial antibiotic-resistance.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animales , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/patogenicidad , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/química , Membrana Celular/efectos de los fármacos , Membrana Celular/microbiología , Resistencia a Medicamentos/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Terapia Molecular Dirigida , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inhibidores , Proteínas Citotóxicas Formadoras de Poros/química , Conformación Proteica , Relación Estructura-Actividad , Virulencia
13.
Int J Biol Macromol ; 178: 253-262, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33636267

RESUMEN

The present study demonstrated that chitin-based nanofibers (CNFs) trigger the chitinase genes (PGIP1 and CaChi2), while elevating salicylic acid that can protect plants against pathogens. Cross-talk between this genetic induction and salicylic-acid-mediated immune response was also observed, which may arm a plant against multiple pathovars. Crab and mushroom based CNFs were synthesized by electrospinning and ball milling techniques. Plants (mung bean, Vigna radiata) (pepper, Capsicum annuum) were pre-inoculated with CNFs and treated with the pathogens Scrolotium rolfsii for pepper and Macrophomina phaseolina for mung bean and shrimp-based CNFs were used as a control. Treated plants had elevated levels of chitinase genes in response to CNFs at inoculation concentrations <10 mg/mL both in soil and media, to protect them against the pathogenic fungal disease. After 24 h of exposure to the pathogens, qRT-PCR showed genes class II chitinase gene (CaChi2) and polygalacturonase inhibitor protein 1 (PGIP1) to be up-regulated in both root and shoot at 0.1 and 1 mg/mL of inoculation, respectively. The ball milled mushroom CNFs were sufficient to trigger the membrane based enzymes with less diameter (≥15 nm) to be most efficient versus others. In vitro analysis showed IC50 of ball milled mushroom CNFs to be most efficient in limiting the growth of fungal biomass. Further trigger-like effects were prominent in reducing pathogenic fungal spread in both species.


Asunto(s)
Ascomicetos/inmunología , Capsicum , Membrana Celular , Quitina , Nanofibras/química , Enfermedades de las Plantas , Inmunidad de la Planta/efectos de los fármacos , Vigna , Capsicum/inmunología , Capsicum/microbiología , Membrana Celular/inmunología , Membrana Celular/microbiología , Quitina/química , Quitina/farmacología , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Vigna/inmunología , Vigna/microbiología
14.
Molecules ; 25(23)2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33291521

RESUMEN

In our previous study, temporin-GHaR (GHaR) showed potent antimicrobial activity with strong hemolytic toxicity. To overcome its weakness, we designed GHaR6R, GHaR7R, GHaR8R, GHaR9R, and GHaR9W by changing the number of positive charges and the hydrophobic surface of GHaR. With the exception of GHaR7R, the hemolytic toxicity of the derived peptides had been reduced, and the antimicrobial activities remained close to the parent peptide (except for GHaR9R). GHaR6R, GHaR7R, GHaR8R, and GHaR9W exhibited a great bactericidal effect on Streptococcus mutans (S. mutans), which is one of the main pathogens causing dental caries. According to the membrane permeation and scanning electron microscope (SEM) analysis, these derived peptides targeted to the cell membranes of planktonic bacteria, contributing to the disruption of the membrane integrity and leakage of the intracellular contents. Moreover, they inhibited the formation of biofilms and eradicated the mature biofilms of S. mutans. Compared with GHaR7R, the derived peptides showed less cytotoxicity to human oral epithelial cells (HOECs). The derived peptides are expected to be the molecular templates for designing antibacterial agents to prevent dental caries.


Asunto(s)
Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Hemólisis/efectos de los fármacos , Péptidos/farmacología , Streptococcus mutans/efectos de los fármacos , Membrana Celular/microbiología , Caries Dental/microbiología , Células Epiteliales/microbiología , Eritrocitos/microbiología , Humanos , Pruebas de Sensibilidad Microbiana/métodos
15.
Genes (Basel) ; 11(12)2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33287151

RESUMEN

Yellow stripe-like (YSL) transporters are required for the transportation of metal-phytosiderophores and are structurally related to metal-nicotianamine complexes. Some studies also reported the involvement of YSL transporters in pathogen-induced defense. However, the molecular mechanisms of YSL genes involved in biotic stress responses are still not clear, especially in cereal crops. This study aimed to functionally characterize TaYS1A during the interaction of wheat and Puccinia striiformis f. sp. tritici (Pst), the causal agent of stripe rust disease. TaYS1A was localized in the cell membrane of wheat protoplasts and Nicotiana benthamiana cells. TaYS1A was significantly up-regulated in wheat leaves after being infected with the avirulent Pst isolate CYR23 and after treatment with salicylic acid (SA). Silencing of TaYS1A by the virus-induced gene silencing method enhanced the susceptibility of wheat to Pst accompanied by reducing the accumulation of SA and H2O2 and down-regulating the transcriptions of TaPR1 and TaPR2. In addition, TaYS1A was found to interact with TaNH2, a homolog of OsNH2, by yeast-two-hybrid assay, and silencing of TaYS1A diminished the expression of TaNH2. Our findings suggested the existence of positive regulation of TaYS1A in providing resistance against Pst by modulating SA-induced signaling and offered new insight into the biological role of YSL in wheat against pathogens.


Asunto(s)
Resistencia a la Enfermedad/genética , Genes de Plantas/genética , Puccinia/patogenicidad , Triticum/genética , Triticum/microbiología , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Membrana Celular/microbiología , Resistencia a la Enfermedad/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/genética , Silenciador del Gen/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/microbiología , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/genética , Hojas de la Planta/microbiología , Protoplastos/efectos de los fármacos , Protoplastos/microbiología , Ácido Salicílico/farmacología , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Nicotiana/efectos de los fármacos , Nicotiana/genética , Nicotiana/microbiología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Triticum/efectos de los fármacos
16.
Elife ; 92020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33226343

RESUMEN

Several virulence lipids populate the outer cell wall of pathogenic mycobacteria. Phthiocerol dimycocerosate (PDIM), one of the most abundant outer membrane lipids, plays important roles in both defending against host antimicrobial programs and in evading these programs altogether. Immediately following infection, mycobacteria rely on PDIM to evade Myd88-dependent recruitment of microbicidal monocytes which can clear infection. To circumvent the limitations in using genetics to understand virulence lipids, we developed a chemical approach to track PDIM during Mycobacterium marinum infection of zebrafish. We found that PDIM's methyl-branched lipid tails enabled it to spread into host epithelial membranes to prevent immune activation. Additionally, PDIM's affinity for cholesterol promoted this phenotype; treatment of zebrafish with statins, cholesterol synthesis inhibitors, decreased spreading and provided protection from infection. This work establishes that interactions between host and pathogen lipids influence mycobacterial infectivity and suggests the use of statins as tuberculosis preventive therapy by inhibiting PDIM spread.


Asunto(s)
Membrana Celular/microbiología , Células Epiteliales/microbiología , Lípidos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/patogenicidad , Factores de Virulencia/metabolismo , Células A549 , Animales , Animales Modificados Genéticamente , Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lípidos/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Estructura Molecular , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Infecciones por Mycobacterium no Tuberculosas/prevención & control , Mycobacterium marinum/efectos de los fármacos , Mycobacterium marinum/genética , Mycobacterium marinum/metabolismo , Relación Estructura-Actividad , Células THP-1 , Virulencia , Factores de Virulencia/química , Pez Cebra
17.
PLoS Pathog ; 16(11): e1009016, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33216805

RESUMEN

The opportunistic pathogen Streptococcus pneumoniae has dual lifestyles: one of an asymptomatic colonizer in the human nasopharynx and the other of a deadly pathogen invading sterile host compartments. The latter triggers an overwhelming inflammatory response, partly driven via pore forming activity of the cholesterol dependent cytolysin (CDC), pneumolysin. Although pneumolysin-induced inflammation drives person-to-person transmission from nasopharynx, the primary reservoir for pneumococcus, it also contributes to high mortality rates, creating a bottleneck that hampers widespread bacterial dissemination, thus acting as a double-edged sword. Serotype 1 ST306, a widespread pneumococcal clone, harbours a non-hemolytic variant of pneumolysin (Ply-NH). Performing crystal structure analysis of Ply-NH, we identified Y150H and T172I as key substitutions responsible for loss of its pore forming activity. We uncovered a novel inter-molecular cation-π interaction, governing formation of the transmembrane ß-hairpins (TMH) in the pore state of Ply, which can be extended to other CDCs. H150 in Ply-NH disrupts this interaction, while I172 provides structural rigidity to domain-3, through hydrophobic interactions, inhibiting TMH formation. Loss of pore forming activity enabled improved cellular invasion and autophagy evasion, promoting an atypical intracellular lifestyle for pneumococcus, a finding that was corroborated in in vivo infection models. Attenuation of inflammatory responses and tissue damage promoted tolerance of Ply-NH-expressing pneumococcus in the lower respiratory tract. Adoption of this altered lifestyle may be necessary for ST306 due to its limited nasopharyngeal carriage, with Ply-NH, aided partly by loss of its pore forming ability, facilitating a benign association of SPN in an alternative, intracellular host niche.


Asunto(s)
Adaptación Fisiológica , Inflamación/microbiología , Mutación con Pérdida de Función , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/fisiología , Estreptolisinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Membrana Celular/microbiología , Colesterol/metabolismo , Citoplasma/microbiología , Femenino , Humanos , Ratones , Modelos Estructurales , Perforina/genética , Perforina/metabolismo , Alineación de Secuencia , Streptococcus pneumoniae/genética , Estreptolisinas/genética
18.
J Microbiol Biotechnol ; 30(12): 1827-1834, 2020 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-33148941

RESUMEN

Candida albicans is a major fungal pathogen in humans. In our previous study, we reported that an ethanol extract from Aucklandia lappa weakens C. albicans cell wall by inhibiting synthesis or assembly of both (1,3)-ß-D-glucan polymers and chitin. In the current study, we found that the extract is involved in permeabilization of C. albicans cell membranes. While uptake of ethidium bromide (EtBr) was 3.0% in control cells, it increased to 7.4% for 30 min in the presence of the A. lappa ethanol extract at its minimal inhibitory concentration (MIC), 0.78 mg/ml, compared to uptake by heat-killed cells. Besides, leakage of DNA and proteins was observed in A. lappa-treated C. albicans cells. The increased uptake of EtBr and leakage of cellular materials suggest that A. lappa ethanol extract induced functional changes in C. albicans cell membranes. Incorporation of diphenylhexatriene (DPH) into membranes in the A. lappa-treated C. albicans cells at its MIC decreased to 84.8%, after 60 min of incubation, compared with that of the controls, indicate that there was a change in membrane dynamics. Moreover, the anticandidal effect of the A. lappa ethanol extract was enhanced at a growth temperature of 40°C compared to that at 35°C. The above data suggest that the antifungal activity of the A. lappa ethanol extract against C. albicans is associated with synergistic action of membrane permeabilization due to changes in membrane dynamics and cell wall damage caused by reduced formation of (1,3)-ß-D-glucan and chitin.


Asunto(s)
Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Saussurea/química , Candidiasis , Membrana Celular/efectos de los fármacos , Membrana Celular/microbiología , Pared Celular/efectos de los fármacos , Quitina/metabolismo , Glucanos/metabolismo , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/farmacología , Temperatura
19.
Microbiology (Reading) ; 166(10): 947-965, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32886602

RESUMEN

Bacterial flagella have many established roles beyond swimming motility. Despite clear evidence of flagella-dependent adherence, the specificity of the ligands and mechanisms of binding are still debated. In this study, the molecular basis of Escherichia coli O157:H7 and Salmonella enterica serovar Typhimurium flagella binding to epithelial cell cultures was investigated. Flagella interactions with host cell surfaces were intimate and crossed cellular boundaries as demarcated by actin and membrane labelling. Scanning electron microscopy revealed flagella disappearing into cellular surfaces and transmission electron microscopy of S. Typhiumurium indicated host membrane deformation and disruption in proximity to flagella. Motor mutants of E. coli O157:H7 and S. Typhimurium caused reduced haemolysis compared to wild-type, indicating that membrane disruption was in part due to flagella rotation. Flagella from E. coli O157 (H7), EPEC O127 (H6) and S. Typhimurium (P1 and P2 flagella) were shown to bind to purified intracellular components of the actin cytoskeleton and directly increase in vitro actin polymerization rates. We propose that flagella interactions with host cell membranes and cytoskeletal components may help prime intimate attachment and invasion for E. coli O157:H7 and S. Typhimurium, respectively.


Asunto(s)
Membrana Celular/microbiología , Citoesqueleto/metabolismo , Escherichia coli O157/fisiología , Flagelos/metabolismo , Salmonella typhimurium/fisiología , Actinas/química , Actinas/metabolismo , Actinas/ultraestructura , Animales , Adhesión Bacteriana , Membrana Celular/metabolismo , Membrana Celular/patología , Membrana Celular/ultraestructura , Células Cultivadas , Citoesqueleto/ultraestructura , Escherichia coli O157/genética , Escherichia coli O157/metabolismo , Flagelos/genética , Flagelos/ultraestructura , Interacciones Huésped-Patógeno , Humanos , Microscopía Electrónica , Mutación , Polimerizacion , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
20.
Adv Exp Med Biol ; 1267: 101-115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32894479

RESUMEN

Pathogenic bacteria colonize or disseminate into cells and tissues by inducing large-scale remodeling of host membranes. The physical phenomena underpinning these massive membrane extension and deformation are poorly understood. Invasive strategies of pathogens have been recently enriched by the description of a spectacular mode of opening of large transendothelial cell macroaperture (TEM) tunnels correlated to the dissemination of EDIN-producing strains of Staphylococcus aureus via a hematogenous route or to the induction of gelatinous edema triggered by the edema toxin from Bacillus anthracis. Remarkably, these highly dynamic tunnels close rapidly after they reach a maximal size. Opening and closure of TEMs in cells lasts for hours without inducing endothelial cell death. Multidisciplinary studies have started to provide a broader perspective of both the molecular determinants controlling cytoskeleton organization at newly curved membranes generated by the opening of TEMs and the physical processes controlling the dynamics of these tunnels. Here we discuss the analogy between the opening of TEM tunnels and the physical principles of dewetting, stemming from a parallel between membrane tension and surface tension. This analogy provides a broad framework to investigate biophysical constraints in cell membrane dynamics and their diversion by certain invasive microbial agents.


Asunto(s)
Bacterias/patogenicidad , Membrana Celular/microbiología , Membrana Celular/patología , Células Endoteliales/microbiología , Células Endoteliales/patología , Humectabilidad , Membrana Celular/metabolismo , Edema/metabolismo , Edema/microbiología , Edema/patología , Células Endoteliales/metabolismo , Humanos , Tensión Superficial
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA