Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 421
Filtrar
1.
Nat Commun ; 15(1): 4740, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834545

RESUMEN

Mitophagy is critical for mitochondrial quality control and function to clear damaged mitochondria. Here, we found that Burkholderia pseudomallei maneuvered host mitophagy for its intracellular survival through the type III secretion system needle tip protein BipD. We identified BipD, interacting with BTB-containing proteins KLHL9 and KLHL13 by binding to the Back and Kelch domains, recruited NEDD8 family RING E3 ligase CUL3 in response to B. pseudomallei infection. Although evidently not involved in regulation of infectious diseases, KLHL9/KLHL13/CUL3 E3 ligase complex was essential for BipD-dependent ubiquitination of mitochondria in mouse macrophages. Mechanistically, we discovered the inner mitochondrial membrane IMMT via host ubiquitome profiling as a substrate of KLHL9/KLHL13/CUL3 complex. Notably, K63-linked ubiquitination of IMMT K211 was required for initiating host mitophagy, thereby reducing mitochondrial ROS production. Here, we show a unique mechanism used by bacterial pathogens that hijacks host mitophagy for their survival.


Asunto(s)
Proteínas Bacterianas , Burkholderia pseudomallei , Macrófagos , Mitocondrias , Mitofagia , Burkholderia pseudomallei/metabolismo , Burkholderia pseudomallei/patogenicidad , Burkholderia pseudomallei/fisiología , Burkholderia pseudomallei/genética , Animales , Ratones , Mitocondrias/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Humanos , Macrófagos/microbiología , Macrófagos/metabolismo , Ubiquitinación , Melioidosis/microbiología , Melioidosis/metabolismo , Interacciones Huésped-Patógeno , Especies Reactivas de Oxígeno/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo III/genética , Ratones Endogámicos C57BL , Membranas Mitocondriales/metabolismo , Células HEK293 , Células RAW 264.7
2.
BMC Genom Data ; 25(1): 47, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783201

RESUMEN

OBJECTIVE: Burkholderia pseudomallei, the etiological cause of melioidosis, is a soil saprophyte endemic in South-East Asia, where it constitutes a public health concern of high-priority. Melioidosis cases are sporadically identified in nonendemic areas, usually associated with travelers or import of goods from endemic regions. Due to extensive intercontinental traveling and the anticipated climate change-associated alterations of the soil bacterial flora, there is an increasing concern for inadvertent establishment of novel endemic areas, which may expand the global burden of melioidosis. Rapid diagnosis, isolation and characterization of B. pseudomallei isolates is therefore of utmost importance particularly in non-endemic locations. DATA DESCRIPTION: We report the genome sequences of two novel clinical isolates (MWH2021 and MST2022) of B. pseudomallei identified in distinct acute cases of melioidosis diagnosed in two individuals arriving to Israel from India and Thailand, respectively. The data includes preliminary genetic analysis of the genomes determining their phylogenetic classification in rapport to the genomes of 131 B. pseudomallei strains documented in the NCBI database. Inspection of the genomic data revealed the presence or absence of loci encoding for several documented virulence determinants involved in the molecular pathogenesis of melioidosis. Virulence analysis in murine models of acute or chronic melioidosis established that both strains belong to the highly virulent class of B. pseudomalleii.


Asunto(s)
Burkholderia pseudomallei , Genoma Bacteriano , Melioidosis , Filogenia , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/aislamiento & purificación , Burkholderia pseudomallei/patogenicidad , Melioidosis/microbiología , Melioidosis/epidemiología , Tailandia/epidemiología , Humanos , Genoma Bacteriano/genética , India , Animales , Israel/epidemiología , Virulencia/genética , Ratones , Secuenciación Completa del Genoma
3.
PLoS Negl Trop Dis ; 16(2): e0010172, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143500

RESUMEN

Burkholderia pseudomallei is a soil-dwelling bacterium endemic to Southeast Asia and northern Australia that causes the disease, melioidosis. Although the global genomic diversity of clinical B. pseudomallei isolates has been investigated, there is limited understanding of its genomic diversity across small geographic scales, especially in soil. In this study, we obtained 288 B. pseudomallei isolates from a single soil sample (~100g; intensive site 2, INT2) collected at a depth of 30cm from a site in Ubon Ratchathani Province, Thailand. We sequenced the genomes of 169 of these isolates that represent 7 distinct sequence types (STs), including a new ST (ST1820), based on multi-locus sequence typing (MLST) analysis. A core genome SNP phylogeny demonstrated that all identified STs share a recent common ancestor that diverged an estimated 796-1260 years ago. A pan-genomics analysis demonstrated recombination between clades and intra-MLST phylogenetic and gene differences. To identify potential differential virulence between STs, groups of BALB/c mice (5 mice/isolate) were challenged via subcutaneous injection (500 CFUs) with 30 INT2 isolates representing 5 different STs; over the 21-day experiment, eight isolates killed all mice, 2 isolates killed an intermediate number of mice (1-2), and 20 isolates killed no mice. Although the virulence results were largely stratified by ST, one virulent isolate and six attenuated isolates were from the same ST (ST1005), suggesting that variably conserved genomic regions may contribute to virulence. Genomes from the animal-challenged isolates were subjected to a bacterial genome-wide association study to identify genomic regions associated with differential virulence. One associated region is a unique variant of Hcp1, a component of the type VI secretion system, which may result in attenuation. The results of this study have implications for comprehensive sampling strategies, environmental exposure risk assessment, and understanding recombination and differential virulence in B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/aislamiento & purificación , Burkholderia pseudomallei/patogenicidad , Melioidosis/microbiología , Filogenia , Microbiología del Suelo , Animales , Burkholderia pseudomallei/clasificación , Burkholderia pseudomallei/genética , Femenino , Genoma Bacteriano , Genómica , Humanos , Ratones Endogámicos BALB C , Tipificación de Secuencias Multilocus , Tailandia , Virulencia
4.
Front Immunol ; 12: 718719, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34456925

RESUMEN

Burkholderia pseudomallei (B. pseudomallei) causes melioidosis, a potentially fatal disease for which no licensed vaccine is available thus far. The host-pathogen interactions in B. pseudomallei infection largely remain the tip of the iceberg. The pathological manifestations are protean ranging from acute to chronic involving one or more visceral organs leading to septic shock, especially in individuals with underlying conditions similar to COVID-19. Pathogenesis is attributed to the intracellular ability of the bacterium to 'step into' the host cell's cytoplasm from the endocytotic vacuole, where it appears to polymerize actin filaments to spread across cells in the closer vicinity. B. pseudomallei effectively evades the host's surveillance armory to remain latent for prolonged duration also causing relapses despite antimicrobial therapy. Therefore, eradication of intracellular B. pseudomallei is highly dependent on robust cellular immune responses. However, it remains ambiguous why certain individuals in endemic areas experience asymptomatic seroconversion, whereas others succumb to sepsis-associated sequelae. Here, we propose key insights on how the host's surveillance radars get commandeered by B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/inmunología , Vigilancia Inmunológica , Melioidosis/inmunología , Animales , Burkholderia pseudomallei/patogenicidad , Interacciones Microbiota-Huesped , Humanos , Virulencia
5.
PLoS Pathog ; 17(5): e1009604, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34048488

RESUMEN

Burkholderia pseudomallei, the etiological agent of melioidosis in humans and animals, often occupies environmental niches and infection sites characterized by limited concentrations of oxygen. Versatile genomic features enable this pathogen to maintain its physiology and virulence under hypoxia, but the crucial regulatory networks employed to switch from oxygen dependent respiration to alternative terminal electron acceptors (TEA) like nitrate, remains poorly understood. Here, we combined a Tn5 transposon mutagenesis screen and an anaerobic growth screen to identify a two-component signal transduction system with homology to RegAB. We show that RegAB is not only essential for anaerobic growth, but also for full virulence in cell lines and a mouse infection model. Further investigations of the RegAB regulon, using a global transcriptomic approach, identified 20 additional regulators under transcriptional control of RegAB, indicating a superordinate role of RegAB in the B. pseudomallei anaerobiosis regulatory network. Of the 20 identified regulators, NarX/L and a FNR homolog were selected for further analyses and a role in adaptation to anaerobic conditions was demonstrated. Growth experiments identified nitrate and intermediates of the denitrification process as the likely signal activateing RegAB, NarX/L, and probably of the downstream regulators Dnr or NsrR homologs. While deletions of individual genes involved in the denitrification process demonstrated their important role in anaerobic fitness, they showed no effect on virulence. This further highlights the central role of RegAB as the master regulator of anaerobic metabolism in B. pseudomallei and that the complete RegAB-mediated response is required to achieve full virulence. In summary, our analysis of the RegAB-dependent modulon and its interconnected regulons revealed a key role for RegAB of B. pseudomallei in the coordination of the response to hypoxic conditions and virulence, in the environment and the host.


Asunto(s)
Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/genética , Melioidosis/microbiología , Adaptación Fisiológica , Anaerobiosis , Animales , Proteínas Bacterianas/genética , Burkholderia pseudomallei/patogenicidad , Burkholderia pseudomallei/fisiología , Femenino , Regulación Bacteriana de la Expresión Génica , Hipoxia , Ratones , Ratones Endogámicos BALB C , Mutación , Nitratos/metabolismo , Oxidación-Reducción , Transcriptoma , Virulencia
6.
Sci Rep ; 11(1): 10405, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34001967

RESUMEN

Burkholderia pseudomallei (Bp) is the causative agent of melioidosis, a disease endemic to the tropics. Melioidosis manifests in various ways ranging from acute skin lesions to pneumonia and, in rare cases, infection of the central nervous system. Bp is a facultative intracellular pathogen and it can infect various cell types. The Bp intracellular lifecycle has been partially elucidated and is highly complex. Herein, we have identified a transcriptional regulator, BP1026B_II1198, that is differentially expressed as Bp transits through host cells. A deletion mutant of BP1026B_II1198 was attenuated in RAW264.7 cell and BALB/c mouse infection. To further characterize the function of this transcriptional regulator, we endeavored to determine the regulon of BP1026B_II1198. RNA-seq analysis showed the global picture of genes regulated while ChIP-seq analysis identified two specific BP1026B_II1198 binding regions on chromosome II. We investigated the transposon mutants of these genes controlled by BP1026B_II1198 and confirmed that these genes contribute to pathogenesis in RAW264.7 murine macrophage cells. Taken together, the data presented here shed light on the regulon of BP1026B_II1198 and its role during intracellular infection and highlights an integral portion of the highly complex regulation network of Bp during host infection.


Asunto(s)
Proteínas Bacterianas/genética , Burkholderia pseudomallei/patogenicidad , Regulación Bacteriana de la Expresión Génica , Melioidosis/microbiología , Proteínas Represoras/genética , Animales , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/genética , Secuenciación de Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Humanos , Ratones , Mutación , Células RAW 264.7 , RNA-Seq , Regulón , Proteínas Represoras/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
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
8.
PLoS Negl Trop Dis ; 15(2): e0009016, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33617546

RESUMEN

Burkholderia pseudomallei is the causative agent of melioidosis, which is a Gram negative, facultative intracellular bacterium. Disease is prevalent in SE Asia and in northern Australia, as well as in other tropical and subtropical regions. Recently, there is an increasing awareness of the importance of bacterial ingestion as a potential route of infection, particularly in cases of unexplained origin of the disease. The marmoset is a New World Monkey (NWM) species that is being developed as an alternative NHP model to complement the more traditionally used Old World Monkeys (OWM). Models have been developed for the traditional routes of disease acquisition, subcutaneous and inhalational. This manuscript details the development and characterisation of an ingestion model of melioidosis. Dose-ranging study assessed the lethality of B. pseudomallei and disease progression was assessed by euthanizing animals at predetermined time points, 12, 36, 48 and 54 hours post-challenge. Challenge doses of greater than 6.2 x 106 cfu resulted in an acute, lethal, febrile disease. Following challenge the lung was the first organ, outside of the gastrointestinal tract, to become colonised. Enteritis (duodenitis, ileitis and/or jejunitis) was observed in sections of the small intestine from animals that succumbed to disease. However, the most severe pathological features were observed in the mesenteric lymph nodes from these animals. These findings are consistent with lymphatic draining as route of dissemination.


Asunto(s)
Burkholderia pseudomallei/fisiología , Sistema Linfático/microbiología , Melioidosis/patología , Animales , Burkholderia pseudomallei/patogenicidad , Callithrix , Modelos Animales de Enfermedad , Enteritis/microbiología , Femenino , Pulmón/microbiología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Masculino , Melioidosis/microbiología
9.
Microb Genom ; 7(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33565959

RESUMEN

Burkholderia pseudomallei, a soil-dwelling Gram-negative bacterium, is the causative agent of the endemic tropical disease melioidosis. Clinical manifestations of B. pseudomallei infection range from acute or chronic localized infection in a single organ to fulminant septicaemia in multiple organs. The diverse clinical manifestations are attributed to various factors, including the genome plasticity across B. pseudomallei strains. We previously characterized B. pseudomallei strains isolated in Malaysia and noted different levels of virulence in model hosts. We hypothesized that the difference in virulence might be a result of variance at the genome level. In this study, we sequenced and assembled four Malaysian clinical B. pseudomallei isolates, UKMR15, UKMPMC2000, UKMD286 and UKMH10. Phylogenomic analysis showed that Malaysian subclades emerged from the Asian subclade, suggesting that the Malaysian strains originated from the Asian region. Interestingly, the low-virulence strain, UKMH10, was the most distantly related compared to the other Malaysian isolates. Genomic island (GI) prediction analysis identified a new island of 23 kb, GI9c, which is present in B. pseudomallei and Burkholderia mallei, but not Burkholderia thailandensis. Genes encoding known B. pseudomallei virulence factors were present across all four genomes, but comparative analysis of the total gene content across the Malaysian strains identified 104 genes that are absent in UKMH10. We propose that these genes may encode novel virulence factors, which may explain the reduced virulence of this strain. Further investigation on the identity and role of these 104 proteins may aid in understanding B. pseudomallei pathogenicity to guide the design of new therapeutics for treating melioidosis.


Asunto(s)
Burkholderia pseudomallei/clasificación , Melioidosis/microbiología , Tipificación de Secuencias Multilocus/métodos , Secuenciación Completa del Genoma/métodos , Técnicas de Tipificación Bacteriana , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/aislamiento & purificación , Burkholderia pseudomallei/patogenicidad , Tamaño del Genoma , Genoma Bacteriano , Islas Genómicas , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Malasia , Filogenia , Factores de Virulencia/genética
10.
ACS Synth Biol ; 10(2): 333-344, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33496568

RESUMEN

Water contamination by pathogenic bacteria is a major public health concern globally. Monitoring bacterial contamination in water is critically important to protect human health, but this remains a critical challenge. Engineered whole-cell biosensors created through synthetic biology hold great promise for rapid and cost-effective detection of waterborne pathogens. In this study, we created a novel whole-cell biosensor to detect water contamination by Pseudomonas aeruginosa and Burkholderia pseudomallei, which are two critical bacterial pathogens and are recognized as common causative agents for waterborne diseases. The biosensor detects the target bacterial pathogens by responding to the relevant quorum sensing signal molecules. Particularly, this study constructed and characterized the biosensor on the basis of the QscR quorum sensing signal system for the first time. We first designed and constructed a QscR on the basis of the sensing module in the E. coli host cell and integrated the QscR sensing module with a reporting module that expressed an enhanced green fluorescent protein (EGFP). The results demonstrated that the biosensor had high sensitivity in response to the quorum sensing signals of the target bacterial pathogens. We further engineered a biosensor that expressed a red pigment lycopene in the reporting module to produce a visible signal readout for the pathogen detection. Additionally, we investigated the feasibility of a paper-based assay by immobilizing the lycopene-based whole-cell biosensor on paper with the aim to build a prototype for developing portable detection devices. The biosensor would provide a simple and inexpensive alternative for timely and point-of-care detection of water contamination and protect human health.


Asunto(s)
Técnicas Biosensibles/métodos , Burkholderia pseudomallei/metabolismo , Regulación Bacteriana de la Expresión Génica , Sistemas de Atención de Punto , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/genética , Microbiología del Agua , Contaminantes del Agua/análisis , Contaminación del Agua/análisis , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidad , Escherichia coli/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Licopeno/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Enfermedades Transmitidas por el Agua/microbiología , Enfermedades Transmitidas por el Agua/prevención & control
11.
Am J Trop Med Hyg ; 104(4): 1260-1264, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432905

RESUMEN

Organisms penetrate the central nervous system (CNS) via three routes. The commonest is the hematogenous route, and other routes include contiguous or penetrating injury or rarely via retrograde axoplasmic route. Although the axoplasmic highway is often used by viruses, only a few bacteria are known to penetrate the CNS via this route. We present a 57-year-old man who developed a penetrating injury while working in a field. Over the next 4 months, he developed pain at the site of the poorly healing wound, which ascended up the right leg and presented as a conus-cauda syndrome. Magnetic resonance imaging (MRI) showed an enhancing intradural intramedullary enhancing lesion in the conus on the right side with cord edema from D11 to L1 level. Extensive evaluation was negative, and he continued to progress to holocord myelitis and developed bilateral corticospinal tract lesions ("tractopathy") in the brain stem and internal capsule. He died after developing a right-sided cerebritis with mass effect. Tissue biopsy from the brain at the time of decompressive craniectomy grew Burkholderia pseudomallei and confirmed a diagnosis of neuromelioidosis (NM). We reviewed the literature for NM, its variable presentations, and the concept of an "infectious tractopathy" and imaging findings which could generate suspicion of this entity.


Asunto(s)
Traumatismos de los Pies/complicaciones , Pie/microbiología , Encefalitis Infecciosa/diagnóstico por imagen , Encefalitis Infecciosa/microbiología , Melioidosis/complicaciones , Mielitis/complicaciones , Antibacterianos/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/microbiología , Burkholderia pseudomallei/patogenicidad , Resultado Fatal , Pie/patología , Traumatismos de los Pies/microbiología , Humanos , Encefalitis Infecciosa/tratamiento farmacológico , Encefalitis Infecciosa/etiología , Imagen por Resonancia Magnética , Masculino , Melioidosis/diagnóstico por imagen , Melioidosis/tratamiento farmacológico , Persona de Mediana Edad , Médula Espinal/patología
12.
Am J Trop Med Hyg ; 104(4): 1247-1251, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432911

RESUMEN

Central nervous system (CNS) melioidosis is a rare neurological infectious disease which carries a high mortality. We describe a previously healthy middle-aged female, who presented to us with left-sided hemiparesis and was on antitubercular therapy from a previous presumed diagnosis of CNS tuberculoma. Non-characteristic imaging picture, multiple negative body fluid cultures, and positive Cerebrospinal fluid galactomannan led to a further delay in diagnosis. Gram stain of the tissue obtained from brain biopsy revealed Gram-negative rods in "safety pin" appearance. By picking up the colonies that appeared on blood agar and MacConkey agar, the identification of the clinical isolates was performed using VITEK® matrix (BioMérieux, Marcy-L'Etoile, France)-assisted laser desorption ionization time-of-flight mass spectrometry (VITEK MALDI TOF MS database version 3.2) which revealed Burkholderia pseudomallei. After the institution of appropriate treatment, she survived but with significant morbidity. A high index of suspicion should be kept for such previously healthy individuals belonging to non-endemic areas, where presentation is suspicious of an infective etiology, but not improving despite appropriate therapy. This may help in early recognition and institution of recommended treatment so that mortality can be avoided.


Asunto(s)
Infecciones Bacterianas del Sistema Nervioso Central/diagnóstico por imagen , Melioidosis/diagnóstico por imagen , Adulto , Antibacterianos/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/microbiología , Encéfalo/patología , Burkholderia pseudomallei/patogenicidad , Infecciones Bacterianas del Sistema Nervioso Central/tratamiento farmacológico , Medios de Cultivo , Femenino , Francia , Humanos , Imagen por Resonancia Magnética , Melioidosis/líquido cefalorraquídeo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Autophagy ; 17(8): 1918-1933, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32777979

RESUMEN

Burkholderia pseudomallei: which causes melioidosis with high mortality in humans, has become a global public health concern. Recently, infection-driven lipid droplet accumulation has been related to the progression of host-pathogen interactions, and its contribution to the pathogenesis of infectious disease has been investigated. Here, we demonstrated that B. pseudomallei infection actively induced a time-dependent increase in the number and size of lipid droplets in human lung epithelial cells and macrophages. We also found that lipid droplet accumulation following B. pseudomallei infection was associated with downregulation of PNPLA2/ATGL (patatin like phospholipase domain containing 2) and lipophagy inhibition. Functionally, lipid droplet accumulation, facilitated via PNPLA2 downregulation, inhibited macroautophagic/autophagic flux and, thus, hindered autophagy-dependent inhibition of B. pseudomallei infection in lung epithelial cells. Mechanistically, we further revealed that nuclear receptor NR1D2 might be involved in the suppression of PNPLA2 after cell exposure to B. pseudomallei. Taken together, our findings unraveled an evolutionary strategy, by which B. pseudomallei interferes with the host lipid metabolism, to block autophagy-dependent suppression of infection. This study proposes potential targets for clinical therapy of melioidosis.Abbreviations: 3-MA: 3-methyladenine; ACTB: actin beta; ATG7: autophagy related 7; B. pseudomallei: Burkholderia pseudomallei; CFU: colony-forming unit; DG: diglyceride; FASN: fatty acid synthase; GFP: green fluorescent protein; LAMP1: lysosomal associated membrane protein 1; LC-MS/MS: liquid chromatography-tandem mass spectrometry; LD: lipid droplet; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MG: monoglyceride; MOI: multiplicity of infection; mRFP: monomeric red fluorescent protein; NR1D2: nuclear receptor subfamily 1 group D member 2; p.i., post-infection; PLIN2/ADRP: perilipin 2; PNPLA2/ATGL: patatin like phospholipase domain containing 2; Rapa: rapamycin; SQSTM1/p62: sequestosome 1; shRNA: short hairpin RNA; TEM: transmission electron microscopy; TG: triglyceride.


Asunto(s)
Autofagia/fisiología , Burkholderia pseudomallei/patogenicidad , Infecciones/tratamiento farmacológico , Lipasa/metabolismo , Metabolismo de los Lípidos/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Humanos , Gotas Lipídicas/metabolismo
14.
Brief Bioinform ; 22(3)2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32444871

RESUMEN

The aerobic, Gram-negative motile bacillus, Burkholderia pseudomallei is a facultative intracellular bacterium causing melioidosis, a critical disease of public health importance, which is widely endemic in the tropics and subtropical regions of the world. Melioidosis is associated with high case fatality rates in animals and humans; even with treatment, its mortality is 20-50%. It also infects plants and is designated as a biothreat agent. B. pseudomallei is pathogenic due to its ability to invade, resist factors in serum and survive intracellularly. Despite its importance, to date only a few effector proteins have been functionally characterized, and there is not much information regarding the host-pathogen protein-protein interactions (PPI) of this system, which are important to studying infection mechanisms and thereby develop prevention measures. We explored two computational approaches, the homology-based interolog and the domain-based method, to predict genome-scale host-pathogen interactions (HPIs) between two different strains of B. pseudomallei (prototypical, and highly virulent) and human. In total, 76 335 common HPIs (between the two strains) were predicted involving 8264 human and 1753 B. pseudomallei proteins. Among the unique PPIs, 14 131 non-redundant HPIs were found to be unique between the prototypical strain and human, compared to 3043 non-redundant HPIs between the highly virulent strain and human. The protein hubs analysis showed that most B. pseudomallei proteins formed a hub with human dnaK complex proteins associated with tuberculosis, a disease similar in symptoms to melioidosis. In addition, drug-binding and carbohydrate-binding mechanisms were found overrepresented within the host-pathogen network, and metabolic pathways were frequently activated according to the pathway enrichment. Subcellular localization analysis showed that most of the pathogen proteins are targeting human proteins inside cytoplasm and nucleus. We also discovered the host targets of the drug-related pathogen proteins and proteins that form T3SS and T6SS in B. pseudomallei. Additionally, a comparison between the unique PPI patterns present in the prototypical and highly virulent strains was performed. The current study is the first report on developing a genome-scale host-pathogen protein interaction networks between the human and B. pseudomallei, a critical biothreat agent. We have identified novel virulence factors and their interacting partners in the human proteome. These PPIs can be further validated by high-throughput experiments and may give new insights on how B. pseudomallei interacts with its host, which will help medical researchers in developing better prevention measures.


Asunto(s)
Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/metabolismo , Simulación por Computador , Melioidosis/metabolismo , Factores de Virulencia/metabolismo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidad , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Melioidosis/tratamiento farmacológico , Melioidosis/genética , Melioidosis/microbiología , Terapia Molecular Dirigida/métodos , Preparaciones Farmacéuticas/administración & dosificación , Unión Proteica/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Virulencia/genética , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/genética
15.
PLoS One ; 15(11): e0241306, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33216758

RESUMEN

Identification of bacterial virulence factors is critical for understanding disease pathogenesis, drug discovery and vaccine development. In this study we used two approaches to predict virulence factors of Burkholderia pseudomallei, the Gram-negative bacterium that causes melioidosis. B. pseudomallei is naturally antibiotic resistant and there are no clinically available melioidosis vaccines. To identify B. pseudomallei protein targets for drug discovery and vaccine development, we chose to search for substrates of the B. pseudomallei periplasmic disulfide bond forming protein A (DsbA). DsbA introduces disulfide bonds into extra-cytoplasmic proteins and is essential for virulence in many Gram-negative organism, including B. pseudomallei. The first approach to identify B. pseudomallei DsbA virulence factor substrates was a large-scale genomic analysis of 511 unique B. pseudomallei disease-associated strains. This yielded 4,496 core gene products, of which we hypothesise 263 are DsbA substrates. Manual curation and database screening of the 263 mature proteins yielded 81 associated with disease pathogenesis or virulence. These were screened for structural homologues to predict potential B-cell epitopes. In the second approach, we searched the B. pseudomallei genome for homologues of the more than 90 known DsbA substrates in other bacteria. Using this approach, we identified 15 putative B. pseudomallei DsbA virulence factor substrates, with two of these previously identified in the genomic approach, bringing the total number of putative DsbA virulence factor substrates to 94. The two putative B. pseudomallei virulence factors identified by both methods are homologues of PenI family ß-lactamase and a molecular chaperone. These two proteins could serve as high priority targets for future B. pseudomallei virulence factor characterization.


Asunto(s)
Proteínas Bacterianas/metabolismo , Vacunas Bacterianas/inmunología , Burkholderia pseudomallei/inmunología , Burkholderia pseudomallei/patogenicidad , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Burkholderia pseudomallei/genética , Cisteína/metabolismo , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Ontología de Genes , Genoma Bacteriano , Modelos Moleculares , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
16.
Sci Rep ; 10(1): 19242, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159122

RESUMEN

Phospholipase C (PLC) enzymes are key virulence factors in several pathogenic bacteria. Burkholderia pseudomallei, the causative agent of melioidosis, possesses at least three plc genes (plc1, plc2 and plc3). We found that in culture medium plc1 gene expression increased with increasing pH, whilst expression of the plc3 gene was pH (4.5 to 9.0) independent. Expression of the plc2 gene was not detected in culture medium. All three plc genes were expressed during macrophage infection by B. pseudomallei K96243. Comparing B. pseudomallei wild-type with plc mutants revealed that plc2, plc12 or plc123 mutants showed reduced intracellular survival in macrophages and reduced plaque formation in HeLa cells. However, plc1 or plc3 mutants showed no significant differences in plaque formation compared to wild-type bacteria. These findings suggest that Plc2, but not Plc1 or Plc3 are required for infection of host cells. In Galleria mellonella, plc1, plc2 or plc3 mutants were not attenuated compared to the wild-type strain, but multiple plc mutants showed reduced virulence. These findings indicate functional redundancy of the B. pseudomallei phospholipases in virulence.


Asunto(s)
Proteínas Bacterianas , Burkholderia pseudomallei , Melioidosis , Fosfolipasas de Tipo C , Factores de Virulencia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/enzimología , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidad , Línea Celular , Melioidosis/enzimología , Melioidosis/genética , Ratones , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
17.
Sci Rep ; 10(1): 17972, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087788

RESUMEN

Tetraspanins are four-span transmembrane proteins of host cells that facilitate infections by many pathogens. Burkholderia pseudomallei is an intracellular bacterium and the causative agent of melioidosis, a severe disease in tropical regions. This study investigated the role of tetraspanins in B. pseudomallei infection. We used flow cytometry to determine tetraspanins CD9, CD63, and CD81 expression on A549 and J774A.1 cells. Their roles in B. pseudomallei infection were investigated in vitro using monoclonal antibodies (MAbs) and recombinant large extracellular loop (EC2) proteins to pretreat cells before infection. Knockout of CD9 and CD81 in cells was performed using CRISPR Cas9 to confirm the role of tetraspanins. Pretreatment of A549 cells with MAb against CD9 and CD9-EC2 significantly enhanced B. pseudomallei internalization, but MAb against CD81 and CD81-EC2 inhibited MNGC formation. Reduction of MNGC formation was consistently observed in J774.A1 cells pretreated with MAbs specific to CD9 and CD81 and with CD9-EC2 and CD81-EC2. Data from knockout experiments confirmed that CD9 enhanced bacterial internalization and that CD81 inhibited MNGC formation. Our data indicate that tetraspanins are host cellular factors that mediated internalization and membrane fusion during B. pseudomallei infection. Tetraspanins may be the potential therapeutic targets for melioidosis.


Asunto(s)
Burkholderia pseudomallei/patogenicidad , Fusión Celular , Melioidosis/microbiología , Fagocitos/fisiología , Tetraspaninas/fisiología , Células A549 , Anticuerpos Monoclonales , Sistemas CRISPR-Cas , Células Cultivadas , Células Gigantes/microbiología , Humanos , Melioidosis/terapia , Tetraspanina 28 , Tetraspanina 29 , Tetraspaninas/metabolismo
18.
Infect Immun ; 89(1)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33106293

RESUMEN

Burkholderia pseudomallei is a Gram-negative bacterium and the causative agent of melioidosis. Despite advances in our understanding of the disease, B. pseudomallei poses a significant health risk, especially in regions of endemicity, where treatment requires prolonged antibiotic therapy. Even though the respiratory and percutaneous routes are well documented and considered the main ways to acquire the pathogen, the gastrointestinal tract is believed to be an underreported and underrecognized route of infection. In the present study, we describe the development of in vitro and in vivo models to study B. pseudomallei gastrointestinal infection. Further, we report that the type 6 secretion system (T6SS) and type 1 fimbriae are important virulence factors required for gastrointestinal infection. Using a human intestinal epithelial cell line and mouse primary intestinal epithelial cells (IECs), we demonstrated that B. pseudomallei adheres, invades, and forms multinucleated giant cells, ultimately leading to cell toxicity. We demonstrated that mannose-sensitive type 1 fimbria is involved in the initial adherence of B. pseudomallei to IECs, although the impact on full virulence was limited. Finally, we also showed that B. pseudomallei requires a functional T6SS for full virulence, bacterial dissemination, and lethality in mice infected by the intragastric route. Overall, we showed that B. pseudomallei is an enteric pathogen and that type 1 fimbria is important for B. pseudomallei intestinal adherence, and we identify a new role for T6SS as a key virulence factor in gastrointestinal infection. These studies highlight the importance of gastrointestinal melioidosis as an understudied route of infection and open a new avenue for the pathogenesis of B. pseudomallei.


Asunto(s)
Burkholderia pseudomallei/fisiología , Gastroenteritis/microbiología , Melioidosis/microbiología , Factores de Virulencia/genética , Animales , Adhesión Bacteriana/genética , Burkholderia pseudomallei/patogenicidad , Modelos Animales de Enfermedad , Fimbrias Bacterianas/fisiología , Regulación Bacteriana de la Expresión Génica , Células Gigantes/microbiología , Células Gigantes/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Sistemas de Secreción Tipo VI , Virulencia/genética
19.
Chem Commun (Camb) ; 56(78): 11528-11547, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32914793

RESUMEN

Trehalose, a disaccharide of glucose, is increasingly recognized as an important contributor to virulence in major bacterial pathogens, such as Mycobacterium tuberculosis, Clostridioides difficile, and Burkholderia pseudomallei. Accordingly, bacterial trehalose metabolic pathways that are not present in humans have gained traction as targets for antibiotic and diagnostic development. Toward this goal, trehalose can be modified through a combination of rational design and synthesis to produce functionalized trehalose analogues, which can be deployed to probe or inhibit bacterial trehalose metabolism. However, the unique α,α-1,1-glycosidic bond and C2 symmetry of trehalose make analogue synthesis via traditional chemical methods very challenging. We and others have turned to the creation of chemoenzymatic synthesis methods, which in principle allow the use of nature's trehalose-synthesizing enzymes to stereo- and regioselectively couple simple, unprotected substrates to efficiently and conveniently generate trehalose analogues. Here, we provide a contextual account of our team's development of a trehalose analogue synthesis method that employs a highly substrate-tolerant, thermostable trehalose synthase enzyme, TreT from Thermoproteus tenax. Then, in three vignettes, we highlight how chemoenzymatic synthesis has accelerated the development of trehalose-based imaging probes and inhibitors that target trehalose-utilizing bacterial pathogens. We describe the role of TreT catalysis and related methods in the development of (i) tools for in vitro and in vivo imaging of mycobacteria, (ii) anti-biofilm compounds that sensitize drug-tolerant mycobacteria to clinical anti-tubercular compounds, and (iii) degradation-resistant trehalose analogues that block trehalose metabolism in C. difficile and potentially other trehalose-utilizing bacteria. We conclude by recapping progress and discussing priorities for future research in this area, including improving the scope and scale of chemoenzymatic synthesis methods to support translational research and expanding the functionality and applicability of trehalose analogues to study and target diverse bacterial pathogens.


Asunto(s)
Burkholderia pseudomallei/efectos de los fármacos , Clostridioides difficile/efectos de los fármacos , Glucosiltransferasas/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Trehalosa/farmacología , Biocatálisis , Biopelículas/efectos de los fármacos , Burkholderia pseudomallei/patogenicidad , Clostridioides difficile/patogenicidad , Clostridioides difficile/fisiología , Farmacorresistencia Bacteriana/efectos de los fármacos , Fluoresceína/química , Mycobacterium tuberculosis/patogenicidad , Thermoproteus/enzimología , Trehalosa/análogos & derivados , Trehalosa/biosíntesis , Virulencia/efectos de los fármacos
20.
PLoS Negl Trop Dis ; 14(9): e0008590, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32991584

RESUMEN

BACKGROUND: Burkholderia pseudomallei is an environmental bacterium that causes melioidosis. A facultative intracellular pathogen, B. pseudomallei can induce multinucleated giant cells (MNGCs) leading to plaque formation in vitro. B. pseudomallei can switch colony morphotypes under stress conditions. In addition, different isolates have been reported to have varying virulence in vivo, but genomic evolution and the relationship with plaque formation is poorly understood. METHODOLOGY/PRINCIPLE FINDINGS: To gain insights into genetic underpinnings of virulence of B. pseudomallei, we screened plaque formation of 52 clinical isolates and 11 environmental isolates as well as 4 isogenic morphotype isolates of B. pseudomallei strains K96243 (types II and III) and 153 (types II and III) from Thailand in A549 and HeLa cells. All isolates except one environmental strain (A4) and K96243 morphotype II were able to induce plaque formation in both cell lines. Intracellular growth assay and confocal microscopy analyses demonstrated that the two plaque-forming-defective isolates were also impaired in intracellular replication, actin polymerization and MNGC formation in infected cells. Whole genome sequencing analysis and PCR revealed that both isolates had a large genomic loss on the same region in chromosome 2, which included Bim cluster, T3SS-3 and T6SS-5 genes. CONCLUSIONS/SIGNIFICANCE: Our plaque screening and genomic studies revealed evidence of impairment in plaque formation in environmental isolates of B. pseudomallei that is associated with large genomic loss of genes important for intracellular multiplication and MNGC formation. These findings suggest that the genomic and phenotypic differences of environmental isolates may be associated with clinical infection.


Asunto(s)
Burkholderia pseudomallei/genética , Burkholderia pseudomallei/aislamiento & purificación , Genoma Bacteriano/genética , Células Gigantes/microbiología , Macrófagos/microbiología , Células A549 , Adulto , Anciano , Burkholderia pseudomallei/patogenicidad , Línea Celular Tumoral , Femenino , Eliminación de Gen , Células HeLa , Humanos , Masculino , Melioidosis/microbiología , Melioidosis/patología , Técnicas Microbiológicas , Persona de Mediana Edad , Estudios Prospectivos , Secuenciación Completa del Genoma , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA