Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Lett Appl Microbiol ; 77(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38331426

RESUMEN

The cariogenicity of Streptococcus mutans relates to its ability to form biofilms on dental surfaces. The aim of this work was to develop a flowcell system compatible with time-lapse confocal microscopy to compare the adhesion and accumulation of S. mutans cells on surfaces in unsupplemented media against media containing sucrose or sucralose (a non-metabolized sweetener) over a short period of time. Fluorescent S. mutans 3209/pVMCherry was suspended in unsupplemented media or media supplemented with 1% sucrose or 1% sucralose and passed through a 3D-printed flowcell system. Flowcells were imaged over 60 minutes using a confocal microscope. Image analysis was performed, including a newly developed object-movement-based method to measure biomass adhesion. Streptococcus mutans 3209/pVMCherry grown in 1% sucrose-supplemented media formed small, dense, relatively immobile clumps in the flowcell system measured by biovolume, surface area, and median object centroid movement. Sucralose-supplemented and un-supplemented media yielded large, loose, mobile aggregates. Architectural metrics and per-object movement were significantly different (P < 0.05) when comparing sucrose-supplemented media to either unsupplemented or sucralose-supplemented media. These results demonstrate the utility of a flowcell system compatible with time-lapse confocal microscopy and image analysis when studying initial biofilm formation and adhesion under different nutritional conditions.


Asunto(s)
Streptococcus mutans , Edulcorantes , Imagen de Lapso de Tiempo , Biopelículas , Sacarosa/farmacología , Microscopía Confocal
2.
Mol Microbiol ; 117(3): 632-660, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34709692

RESUMEN

The use of soluble fusion proteins of pattern recognition receptors (PRRs) used in the detection of exogenous and endogenous ligands has helped resolve the roles of PRRs in the innate immune response to pathogens, how they shape the adaptive immune response, and function in maintaining homeostasis. Using the immunoglobulin (Ig) crystallizable fragment (Fc) domain as a fusion partner, the PRR fusion proteins are soluble, stable, easily purified, have increased affinity due to the Fc homodimerization properties, and consequently have been used in a wide range of applications such as flow cytometry, screening of protein and glycan arrays, and immunofluorescent microscopy. This review will predominantly focus on the recognition of pathogens by the cell membrane-expressed glycan-binding proteins of the C-type lectin receptor (CLR) subgroup of PRRs. PRRs bind to conserved pathogen-associated molecular patterns (PAMPs), such as glycans, usually located within or on the outer surface of the pathogen. Significantly, many glycans structures are identical on both host and pathogen (e.g. the Lewis (Le) X glycan), allowing the use of Fc CLR fusion proteins with known endogenous and/or exogenous ligands as tools to identify pathogen structures that are able to interact with the immune system. Screens of highly purified pathogen-derived cell wall components have enabled identification of many unique PAMP structures recognized by CLRs. This review highlights studies using Fc CLR fusion proteins, with emphasis on the PAMPs found in fungi, bacteria, viruses, and parasites. The structure and unique features of the different CLR families is presented using examples from a broad range of microbes whenever possible.


Asunto(s)
Lectinas Tipo C , Moléculas de Patrón Molecular Asociado a Patógenos , Interacciones Huésped-Patógeno , Humanos , Lectinas Tipo C/metabolismo , Ligandos , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo
3.
Cell Microbiol ; 23(2): e13275, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33006213

RESUMEN

The peritrophic matrix (PM) is an acellular membrane that covers the gut epithelium in arthropods and physically separates it from the lumen. The structure is thought to play an important role in tick biology. The PM is also known to impact the persistence of tick-borne pathogens like Borrelia burgdorferi, although limited information is available about its molecular constituents or their biological significance. Herein, we characterise a novel PM-associated gut protein in Ixodes scapularis ticks, annotated as Peritrophic Membrane Chitin Binding Protein (PM_CBP), for its role in the integrity and function of the matrix. The PM_CBP displays homology to the chitin deacetylase metalloenzyme, shows upregulation during tick feeding, and is localized at the luminal surface of the gut epithelium. The structural integrity of the PM was impaired both by the knock down of PM_CBP expression via RNA interference and by treatment with anti-PM_CBP antibodies, as revealed by its electron microscopic appearance. Additionally, the duration of tick engorgement on mice and the passage of experimentally-inoculated fluorescent dextran molecules across the PM are affected by the knock down of PM_CBP expression. The transfer of anti-PM_CBP antibodies into the tick gut impacted the overall composition of the resident microbiome, and also influenced B. burgdorferi acquisition in ticks and its transmission to mice. Taken together, these data highlight the biological significance of the Ixodes PM and suggest that the targeting of its molecular constituents may contribute to the development of novel interventions against tick-borne infections.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Borrelia burgdorferi/fisiología , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Ixodes/metabolismo , Ixodes/microbiología , Enfermedad de Lyme/microbiología , Animales , Borrelia burgdorferi/patogenicidad , Proteínas Portadoras/metabolismo , Quitina/metabolismo , ADN Bacteriano , Femenino , Técnicas de Silenciamiento del Gen , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C3H , Unión Proteica , Interferencia de ARN , ARN Ribosómico 16S
4.
Cell Microbiol ; 23(9): e13366, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34021690

RESUMEN

Many enteric pathogens employ a type III secretion system (T3SS) to translocate effector proteins directly into the host cell cytoplasm, where they subvert signalling pathways of the intestinal epithelium. Here, we report that the anti-apoptotic regulator HS1-associated protein X1 (HAX-1) is an interaction partner of the T3SS effectors EspO of enterohaemorrhagic Escherichia coli (EHEC) and Citrobacter rodentium, OspE of Shigella flexneri and Osp1STYM of Salmonella enterica serovar Typhimurium. EspO, OspE and Osp1STYM have previously been reported to interact with the focal adhesions protein integrin linked kinase (ILK). We found that EspO localizes both to the focal adhesions (ILK localisation) and mitochondria (HAX-1 localisation), and that increased expression of HAX-1 leads to enhanced mitochondrial localisation of EspO. Ectopic expression of EspO, OspE and Osp1STYM protects cells from apoptosis induced by staurosporine and tunicamycin. Depleting cells of HAX-1 indicates that the anti-apoptotic activity of EspO is HAX-1 dependent. Both HAX-1 and ILK were further confirmed as EspO1-interacting proteins during infection using T3SS-delivered EspO1. Using cell detachment as a proxy for cell death we confirmed that T3SS-delivered EspO1 could inhibit cell death induced during EPEC infection, to a similar extent as the anti-apoptotic effector NleH, or treatment with the pan caspase inhibitor z-VAD. In contrast, in cells lacking HAX-1, EspO1 was no longer able to protect against cell detachment, while NleH1 and z-VAD maintained their protective activity. Therefore, during both infection and ectopic expression EspO protects cells from cell death by interacting with HAX-1. These results suggest that despite the differences between EHEC, C. rodentium, Shigella and S. typhimurium infections, hijacking HAX-1 anti-apoptotic signalling is a common strategy to maintain the viability of infected cells. TAKE AWAY: EspO homologues are found in EHEC, Shigella, S. typhimurium and some EPEC. EspO homologues interact with HAX-1. EspO protects infected cells from apoptosis. EspO joins a growing list of T3SS effectors that manipulate cell death pathways.


Asunto(s)
Escherichia coli Enterohemorrágica , Escherichia coli Enteropatógena , Proteínas de Escherichia coli , Apoptosis , Citrobacter rodentium , Sistemas de Secreción Tipo III
5.
Cell Microbiol ; 23(7): e13337, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33813790

RESUMEN

Intracellular pathogens interact with host systems in intimate ways to sustain a pathogenic lifestyle. Consequently, these interactions can potentially be targets of host-directed interventions against infectious diseases. In case of tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis (Mtb), while effective anti-tubercular compounds are available, the long treatment duration and emerging drug resistance necessitate identification of new class of molecules with anti-TB activity, as well as new treatment strategies. A significant part of the effort in finding new anti-TB drugs is focused on bacterial targets in bacterial systems. However, the host environment plays a major role in pathogenesis mechanisms and must be considered actively in these efforts. On the one hand, the bacterial origin targets must be relevant and accessible in the host, while on the other hand, new host origin targets required for the bacterial survival can be targeted. Such targets are good candidates for host-directed therapeutics, a strategy gaining traction as an adjunct in TB treatment. In this review, we will summarise the screening platforms used to identify compounds with anti-tubercular activities inside different host environments and outline recent technical advances in these platforms. Finally, while the examples given are specific to mycobacteria, the methods and principles outlined are broadly applicable to most intracellular infections.


Asunto(s)
Antituberculosos , Evaluación Preclínica de Medicamentos/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Humanos
6.
J Appl Microbiol ; 132(4): 3302-3314, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34919308

RESUMEN

AIMS: Mycobacterium abscessus subsp. abscessus (MABS) is an emerging, opportunistic pathogen found globally in freshwater biofilms and soil. Typically, isolates are treated as a uniform group of organisms and very little is known about their comparative survival in healthy host cells. We posit that environmentally- and clinically derived isolates, show differential infectivity in immune cells and resistance to innate defenses. METHODS AND RESULTS: Six MABS isolates were tested including three water biofilm/soil and three sputum-derived isolates. A clinical MABS type strain and an environmental isolate of Arthrobacter were also included. MABS counts were significantly higher compared to Arthrobacter after co-culture with Acanthamoeba lenticulata, BEAS-2B epithelial cells, alveolar macrophages and the THP-1 macrophage cell line. A rough sputum-derived MABS isolate emerged as an isolate with higher virulence compared to others tested, as both a pellicle and cord former, survivor in the human cell models tested, inducer of high and prolonged production of pro-inflammatory cytokines, and the capacity to evade LL-37. CONCLUSIONS: Findings support intraspecies variation between MABS isolates. SIGNIFICANCE AND IMPACT OF THE STUDY: These data indicate subversion of host immune defenses by environmental and clinical MABS isolates is nuanced and maybe isolate dependent, providing new information regarding the pathogenesis of NTM infections.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium abscessus , Biopelículas , Humanos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Micobacterias no Tuberculosas/genética , Esputo , Virulencia
7.
Cell Microbiol ; 22(4): e13187, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32185897

RESUMEN

Pioneer work by Prof. Cossart among others, studying the interactions between pathogenic bacteria and host cells (this discipline was termed Cellular Microbiology), was fundamental to determine the bacterial infection processes and to improve our knowledge of different cellular mechanisms. The study of bacteria-host interactions also involves in vivo host immune responses, which can be manipulated by bacteria, being these last potent tools for different immunotherapies. During the last years, tumour immunotherapies, mainly the use of antibodies that target immune checkpoints [checkpoint inhibitors (CPI)], have been a revolution in oncology, allowing the treatment of tumours otherwise with very bad prognosis. In the same direction, bacteria inoculations have been used from long to treat some cancers; for example, non-muscle-invasive bladder cancer can be successfully treated with the bacterium Bacillus Calmette Guerin (BCG). More recently, it has been shown that microbiota could determine the success of CPI immunotherapies and intense research is being performed in order to use bacteria as immunotherapy tools due to their ability to activate the immune system. In this context, to expand the knowledge of the bacteria-immune system interactions will be fundamental to improve tumour immunotherapies.


Asunto(s)
Bacterias/inmunología , Interacciones Microbiota-Huesped/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Ensayos Clínicos como Asunto , Humanos , Mycobacterium bovis/inmunología
8.
Cell Microbiol ; 22(11): e13251, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32779854

RESUMEN

During acute Pseudomonas aeruginosa infection, the inflammatory response is essential for bacterial clearance. Neutrophil recruitment can be initiated following the assembly of an inflammasome within sentinel macrophages, leading to activation of caspase-1, which in turn triggers macrophage pyroptosis and IL-1ß/IL-18 maturation. Inflammasome formation can be induced by a number of bacterial determinants, including Type III secretion systems (T3SSs) or pore-forming toxins, or, alternatively, by lipopolysaccharide (LPS) via caspase-11 activation. Surprisingly, previous studies indicated that a T3SS-induced inflammasome increased pathogenicity in mouse models of P. aeruginosa infection. Here, we investigated the immune reaction of mice infected with a T3SS-negative P. aeruginosa strain (IHMA879472). Virulence of this strain relies on ExlA, a secreted pore-forming toxin. IHMA879472 promoted massive neutrophil infiltration in infected lungs, owing to efficient priming of toll-like receptors, and thus enhanced the expression of inflammatory proteins including pro-IL-1ß and TNF-α. However, mature-IL-1ß and IL-18 were undetectable in wild-type mice, suggesting that ExlA failed to effectively activate caspase-1. Nevertheless, caspase-1/11 deficiency improved survival following infection with IHMA879472, as previously described for T3SS+ bacteria. We conclude that the detrimental effect associated with the ExlA-induced inflammasome is probably not due to hyperinflammation, rather it stems from another inflammasome-dependent process.


Asunto(s)
Inflamasomas/inmunología , Leucocidinas/toxicidad , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Animales , Citocinas/biosíntesis , Inflamasomas/metabolismo , Inflamación , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Infiltración Neutrófila , Fragmentos de Péptidos/metabolismo , Pseudomonas aeruginosa/crecimiento & desarrollo , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/metabolismo , Sistemas de Secreción Tipo III , Virulencia
9.
Cell Microbiol ; 22(8): e13210, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32329205

RESUMEN

Campylobacter jejuni is the leading cause of bacterial-derived gastroenteritis worldwide and can lead to several post-infectious inflammatory disorders. Despite the prevalence and health impacts of the bacterium, interactions between the host innate immune system and C. jejuni remain poorly understood. To expand on earlier work demonstrating that neutrophils traffic to the site of infection in an animal model of campylobacteriosis, we identified significant increases in several predominantly neutrophil-derived proteins in the faeces of C. jejuni-infected patients, including lipocalin-2, myeloperoxidase and neutrophil elastase. In addition to demonstrating that these proteins significantly inhibited C. jejuni growth, we determined they are released during formation of C. jejuni-induced neutrophil extracellular traps (NETs). Using quantitative and qualitative methods, we found that purified human neutrophils are activated by C. jejuni and exhibit signatures of NET generation, including presence of protein arginine deiminase-4, histone citrullination, myeloperoxidase, neutrophil elastase release and DNA extrusion. Production of NETs correlated with C. jejuni phagocytosis/endocytosis and invasion of neutrophils suggesting that host- and bacterial-mediated activities are responsible for NET induction. Further, NET-like structures were observed within intestinal tissue of C. jejuni-infected ferrets. Finally, induction of NETs significantly increased human colonocyte cytotoxicity, indicating that NET formation during C. jejuni infection may contribute to observed tissue pathology. These findings provide further understanding of C. jejuni-neutrophil interactions and inflammatory responses during campylobacteriosis.


Asunto(s)
Campylobacter jejuni/inmunología , Campylobacter jejuni/fisiología , Trampas Extracelulares/inmunología , Trampas Extracelulares/microbiología , Heces/química , Interacciones Microbiota-Huesped/inmunología , Neutrófilos/inmunología , Animales , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/microbiología , Células Cultivadas , Colon/citología , Colon/microbiología , Colon/patología , Hurones , Humanos , Inflamación , Elastasa de Leucocito/metabolismo , Masculino , Neutrófilos/química , Neutrófilos/microbiología , Fagocitosis
10.
Cell Microbiol ; 22(7): e13194, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32068949

RESUMEN

Cytolethal distending toxins (Cdt) are a family of toxins produced by several human pathogens which infect mucocutaneous tissue and induce inflammatory disease. We have previously demonstrated that the Aggregatibacter actinomycetemcomitans Cdt induces a pro-inflammatory response from human macrophages which involves activation of the NLRP3 inflammasome. We now demonstrate that in addition to activating caspase-1 (canonical inflammasome), Cdt treatment leads to caspase-4 activation and involvement of the noncanonical inflammasome. Cdt-treated cells exhibit pyroptosis characterised by cleavage of gasdermin-D (GSDMD), release of HMGB1 at 24 hr and LDH at 48 hr. Inhibition of either the canonical (caspase-1) or noncanonical (caspase-4) inflammasome blocks both Cdt-induced release of IL-1ß and induction of pyroptosis. Analysis of upstream events indicates that Cdt induces Syk phosphorylation (activation); furthermore, blockade of Syk expression and inhibition of pSyk activity inhibit both Cdt-induced cytokine release and pyroptosis. Finally, we demonstrate that increases in pSyk are dependent upon Cdt-induced activation of GSK3ß. These studies advance our understanding of Cdt function and provide new insight into the virulence potential of Cdt in mediating the pathogenesis of disease caused by Cdt-producing organisms such as A. actinomycetemcomitans.


Asunto(s)
Toxinas Bacterianas/efectos adversos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Quinasa Syk/metabolismo , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Citocinas/metabolismo , Proteína HMGB1/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis , Células THP-1
11.
Cell Microbiol ; 22(1): e13126, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31610608

RESUMEN

The mouse pathogen Citrobacter rodentium is used to model infections with enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC). Pathogenesis is commonly modelled in mice developing mild disease (e.g., C57BL/6). However, little is known about host responses in mice exhibiting severe colitis (e.g., C3H/HeN), which arguably provide a more clinically relevant model for human paediatric enteric infection. Infection of C3H/HeN mice with C. rodentium results in rapid colonic colonisation, coinciding with induction of key inflammatory signatures and colonic crypt hyperplasia. Infection also induces dramatic changes to bioenergetics in intestinal epithelial cells, with transition from oxidative phosphorylation (OXPHOS) to aerobic glycolysis and higher abundance of SGLT4, LDHA, and MCT4. Concomitantly, mitochondrial proteins involved in the TCA cycle and OXPHOS were in lower abundance. Similar to observations in C57BL/6 mice, we detected simultaneous activation of cholesterol biogenesis, import, and efflux. Distinctly, however, the pattern recognition receptors NLRP3 and ALPK1 were specifically induced in C3H/HeN. Using cell-based assays revealed that C. rodentium activates the ALPK1/TIFA axis, which is dependent on the ADP-heptose biosynthesis pathway but independent of the Type III secretion system. This study reveals for the first time the unfolding intestinal epithelial cells' responses during severe infectious colitis, which resemble EPEC human infections.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interacciones Microbiota-Huesped , Inflamación/microbiología , Mucosa Intestinal/microbiología , Animales , Citrobacter rodentium/patogenicidad , Colitis/inmunología , Colitis/microbiología , Infecciones por Enterobacteriaceae/metabolismo , Femenino , Microbioma Gastrointestinal , Células HeLa , Humanos , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteómica , Organismos Libres de Patógenos Específicos
12.
Cell Microbiol ; 22(1): e13128, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31652371

RESUMEN

Leprosy neuropathy is a chronic degenerative infectious disorder of the peripheral nerve caused by the intracellular obligate pathogen Mycobacterium leprae (M. leprae). Among all nonneuronal cells that constitute the nerve, Schwann cells are remarkable in supporting M. leprae persistence intracellularly. Notably, the success of leprosy infection has been attributed to its ability in inducing the demyelination phenotype after contacting myelinated fibres. However, the exact role M. leprae plays during the ongoing process of myelin breakdown is entirely unknown. Here, we provided evidence showing an unexpected predilection of leprosy pathogen for degenerating myelin ovoids inside Schwann cells. In addition, M. leprae infection accelerated the rate of myelin breakdown and clearance leading to increased formation of lipid droplets, by modulating a set of regulatory genes involved in myelin maintenance, autophagy, and lipid storage. Remarkably, the blockage of myelin breakdown significantly reduced M. leprae content, demonstrating a new unpredictable role of myelin dismantling favouring M. leprae physiology. Collectively, our study provides novel evidence that may explain the demyelination phenotype as an evolutionarily conserved mechanism used by leprosy pathogen to persist longer in the peripheral nerve.


Asunto(s)
Mycobacterium leprae/fisiología , Vaina de Mielina/metabolismo , Células de Schwann/microbiología , Animales , Células Cultivadas , Humanos , Lepra/complicaciones , Lepra/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Mycobacterium leprae/patogenicidad , Vaina de Mielina/microbiología
13.
Cell Microbiol ; 22(2): e13141, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31709673

RESUMEN

Extracellular adenosine production is crucial for host resistance against Streptococcus pneumoniae (pneumococcus) and is thought to affect antibacterial immune responses by neutrophils. However, whether extracellular adenosine alters direct host-pathogen interaction remains unexplored. An important determinant for lung infection by S. pneumoniae is its ability to adhere to the pulmonary epithelium. Here we explored whether extracellular adenosine can directly impact bacterial adherence to lung epithelial cells. We found that signaling via A1 adenosine receptor significantly reduced the ability of pneumococci to bind human pulmonary epithelial cells. A1 receptor signaling blocked bacterial binding by reducing the expression of platelet-activating factor receptor, a host protein used by S. pneumoniae to adhere to host cells. In vivo, A1 was required for control of pneumococcal pneumonia as inhibiting it resulted in increased host susceptibility. As S. pneumoniae remain a leading cause of community-acquired pneumonia in the elderly, we explored the role of A1 in the age-driven susceptibility to infection. We found no difference in A1 pulmonary expression in young versus old mice. Strikingly, triggering A1 signaling boosted host resistance of old mice to S. pneumoniae pulmonary infection. This study demonstrates a novel mechanism by which extracellular adenosine modulates resistance to lung infection by targeting bacterial-host interactions.


Asunto(s)
Células Epiteliales/microbiología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Neumonía Neumocócica , Receptor de Adenosina A1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Streptococcus pneumoniae , Factores de Edad , Animales , Adhesión Bacteriana , Línea Celular , Células Epiteliales/citología , Células Epiteliales/inmunología , Interacciones Huésped-Patógeno , Humanos , Pulmón/citología , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Streptococcus pneumoniae/inmunología
14.
Cell Microbiol ; 21(10): e13084, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31290228

RESUMEN

Toxoplasma gondii causes retinitis and encephalitis. Avoiding targeting by autophagosomes is key for its survival because T. gondii cannot withstand lysosomal degradation. During invasion of host cells, T. gondii triggers epidermal growth factor receptor (EGFR) signalling enabling the parasite to avoid initial autophagic targeting. However, autophagy is a constitutive process indicating that the parasite may also use a strategy operative beyond invasion to maintain blockade of autophagic targeting. Finding that such a strategy exists would be important because it could lead to inhibition of host cell signalling as a novel approach to kill the parasite in previously infected cells and treat toxoplasmosis. We report that T. gondii induced prolonged EGFR autophosphorylation. This effect was mediated by PKCα/PKCß âž” Src because T. gondii caused prolonged activation of these molecules and their knockdown or incubation with inhibitors of PKCα/PKCß or Src after host cell invasion impaired sustained EGFR autophosphorylation. Addition of EGFR tyrosine kinase inhibitor (TKI) to previously infected cells led to parasite entrapment by LC3 and LAMP-1 and pathogen killing dependent on the autophagy proteins ULK1 and Beclin 1 as well as lysosomal enzymes. Administration of gefitinib (EGFR TKI) to mice with ocular and cerebral toxoplasmosis resulted in disease control that was dependent on Beclin 1. Thus, T. gondii promotes its survival through sustained EGFR signalling driven by PKCα/ß âž” Src, and inhibition of EGFR controls pre-established toxoplasmosis.


Asunto(s)
Autofagosomas/metabolismo , Autofagosomas/parasitología , Autofagia , Receptores ErbB/metabolismo , Toxoplasmosis Animal/tratamiento farmacológico , Toxoplasmosis Animal/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/enzimología , Autofagia/efectos de los fármacos , Autofagia/genética , Beclina-1/metabolismo , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Femenino , Gefitinib/uso terapéutico , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Fosforilación , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Toxoplasma/efectos de los fármacos , Toxoplasma/patogenicidad , Toxoplasmosis Animal/enzimología , Toxoplasmosis Animal/genética
15.
Cell Microbiol ; 21(9): e13046, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31099152

RESUMEN

The virulence strategy of pathogenic Yersinia spp. involves cell-invasive as well as phagocytosis-preventing tactics to enable efficient colonisation of the host organism. Enteropathogenic yersiniae display an invasive phenotype in early infection stages, which facilitates penetration of the intestinal mucosa. Here we show that invasion of epithelial cells by Yersinia enterocolitica is followed by intracellular survival and multiplication of a subset of ingested bacteria. The replicating bacteria were enclosed in vacuoles with autophagy-related characteristics, showing phagophore formation, xenophagy, and recruitment of cytoplasmic autophagosomes to the bacteria-containing compartments. The subsequent fusion of these vacuoles with lysosomes and concomitant vesicle acidification were actively blocked by Yersinia. This resulted in increased intracellular proliferation and detectable egress of yersiniae from infected cells. Notably, deficiency of the core autophagy machinery component FIP200 impaired the development of autophagic features at Yersinia-containing vacuoles as well as intracellular replication and release of bacteria to the extracellular environment. These results suggest that Y. enterocolitica may take advantage of the macroautophagy pathway in epithelial cells to create an autophagosomal niche that supports intracellular bacterial survival, replication, and, eventually, spread of the bacteria from infected cells.


Asunto(s)
Autofagosomas/microbiología , Células Epiteliales/microbiología , Yersinia enterocolitica/patogenicidad , Animales , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Muerte Celular , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Células HeLa , Interacciones Microbiota-Huesped , Humanos , Lisosomas/metabolismo , Lisosomas/microbiología , Lisosomas/ultraestructura , Ratones , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Vacuolas/metabolismo , Vacuolas/microbiología , Vacuolas/ultraestructura , Yersinia enterocolitica/crecimiento & desarrollo , Yersinia enterocolitica/metabolismo
16.
Cell Microbiol ; 21(1): e12971, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30370624

RESUMEN

Legionella pneumophila is a ubiquitous environmental bacterium that has evolved to infect and proliferate within amoebae and other protists. It is thought that accidental inhalation of contaminated water particles by humans is what has enabled this pathogen to proliferate within alveolar macrophages and cause pneumonia. However, the highly evolved macrophages are equipped with more sophisticated innate defence mechanisms than are protists, such as the evolution of phagotrophic feeding into phagocytosis with more evolved innate defence processes. Not surprisingly, the majority of proteins involved in phagosome biogenesis (~80%) have origins in the phagotrophy stage of evolution. There are a plethora of highly evolved cellular and innate metazoan processes, not represented in protist biology, that are modulated by L. pneumophila, including TLR2 signalling, NF-κB, apoptotic and inflammatory processes, histone modification, caspases, and the NLRC-Naip5 inflammasomes. Importantly, L. pneumophila infects haemocytes of the invertebrate Galleria mellonella, kill G. mellonella larvae, and proliferate in and kill Drosophila adult flies and Caenorhabditis elegans. Although coevolution with protist hosts has provided a substantial blueprint for L. pneumophila to infect macrophages, we discuss the further evolutionary aspects of coevolution of L. pneumophila and its adaptation to modulate various highly evolved innate metazoan processes prior to becoming a human pathogen.


Asunto(s)
Amoeba/metabolismo , Amoeba/microbiología , Evasión Inmune , Inmunidad Innata , Legionella pneumophila/fisiología , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Caenorhabditis elegans/inmunología , Caenorhabditis elegans/microbiología , Drosophila/inmunología , Drosophila/microbiología , Lepidópteros/inmunología , Lepidópteros/microbiología
17.
Cell Microbiol ; 21(1): e12958, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30251327

RESUMEN

Professional phagocytic cells such as macrophages are a central part of innate immune defence. They ingest microorganisms into membrane-bound compartments (phagosomes), which acidify and eventually fuse with lysosomes, exposing their contents to a microbicidal environment. Gram-positive Rhodococcus equi can cause pneumonia in young foals and in immunocompromised humans. The possession of a virulence plasmid allows them to subvert host defence mechanisms and to multiply in macrophages. Here, we show that the plasmid-encoded and secreted virulence-associated protein A (VapA) participates in exclusion of the proton-pumping vacuolar-ATPase complex from phagosomes and causes membrane permeabilisation, thus contributing to a pH-neutral phagosome lumen. Using fluorescence and electron microscopy, we show that VapA is also transferred from phagosomes to lysosomes where it permeabilises the limiting membranes for small ions such as protons. This permeabilisation process is different from that of known membrane pore formers as revealed by experiments with artificial lipid bilayers. We demonstrate that, at 24 hr of infection, virulent R. equi is contained in a vacuole, which is enriched in lysosome material, yet possesses a pH of 7.2 whereas phagosomes containing a vapA deletion mutant have a pH of 5.8 and those with virulence plasmid-less sister strains have a pH of 5.2. Experimentally neutralising the macrophage endocytic system allows avirulent R. equi to multiply. This observation is mirrored in the fact that virulent and avirulent R. equi multiply well in extracts of purified lysosomes at pH 7.2 but not at pH 5.1. Together these data indicate that the major function of VapA is to generate a pH-neutral and hence growth-promoting intracellular niche. VapA represents a new type of Gram-positive virulence factor by trafficking from one subcellular compartment to another, affecting membrane permeability, excluding proton-pumping ATPase, and consequently disarming host defences.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Fagosomas/microbiología , ATPasas de Translocación de Protón/antagonistas & inhibidores , Rhodococcus equi/crecimiento & desarrollo , Rhodococcus equi/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Ratones , Microscopía Electrónica , Microscopía Fluorescente , Virulencia
18.
Cell Microbiol ; 21(2): e12992, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30550623

RESUMEN

Lyme arthritis (LA), a late disease manifestation of Borrelia burgdorferi infection, usually resolves with antibiotic therapy. However, some patients develop proliferative synovitis lasting months to several years after spirochetal killing, called postinfectious LA. In this study, we phenotyped haematopoietic and stromal cell populations in the synovial lesion ex vivo and used these findings to generate an in vitro model of LA using patient-derived fibroblast-like synoviocytes (FLS). Ex vivo analysis of synovial tissue revealed high abundance of IFNγ-producing T cells and NK cells. Similar to marked IFNγ responses in tissue, postinfectious LA synovial fluid also had high levels of IFNγ. HLA-DR-positive FLS were present throughout the synovial lesion, particularly in areas of inflammation. FLS stimulated in vitro with B. burgdorferi, which were similar to conditions during infection, expressed 68 genes associated primarily with innate immune activation and neutrophil recruitment. In contrast, FLS stimulated with IFNγ, which were similar to conditions in the postinfectious phase, expressed >2,000 genes associated with pathogen sensing, inflammation, and MHC Class II antigen presentation, similar to the expression profile in postinfectious synovial tissue. Furthermore, costimulation of FLS with B. burgdorferi and IFNγ induced greater expression of IL-6 and other innate immune response proteins and genes than with IFNγ stimulation alone. These results suggest that B. burgdorferi infection, in combination with IFNγ, initiates the differentiation of FLS into a highly inflammatory phenotype. We hypothesise that overexpression of IFNγ by lymphocytes within synovia perpetuates these responses in the postinfectious period, causing proliferative synovitis and stalling appropriate repair of damaged tissue.


Asunto(s)
Fibroblastos/citología , Interferón gamma/inmunología , Enfermedad de Lyme/inmunología , Sinoviocitos/citología , Sinovitis/inmunología , Borrelia burgdorferi/inmunología , Diferenciación Celular/inmunología , Humanos , Enfermedad de Lyme/patología , Membrana Sinovial/metabolismo , Linfocitos T/inmunología
19.
Cell Microbiol ; 21(11): e13079, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31265745

RESUMEN

Inflammasomes are cytosolic, multimeric protein complexes capable of activating pro-inflammatory cytokines such as IL-1ß and IL-18, which play a key role in host defence. Inflammasome components are highly expressed in the intestinal epithelium. In recent years, studies have begun to demonstrate that epithelial-intrinsic inflammasomes play a critical role in regulating epithelial homeostasis, both by defending the epithelium from pathogenic insult and through the regulation of the mucosal environment. However, the majority of research regarding inflammasome activation has focused on professional immune cells, such as macrophages. Here, we present an overview of the current understanding of inflammasome function in epithelial cells and at mucosal surfaces and, in particular, in the intestine.


Asunto(s)
Células Epiteliales/metabolismo , Inflamasomas/metabolismo , Mucosa Intestinal/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Regulación de la Expresión Génica/inmunología , Interacciones Microbiota-Huesped , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Microbiota/inmunología , Interleucina-22
20.
Cell Microbiol ; 21(10): e13074, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31231933

RESUMEN

Cholera toxin (Ctx) is an AB-type protein toxin that acts as an adenosine diphosphate (ADP)-ribosyltransferase to disrupt intracellular signalling in the target cell. It moves by vesicle carriers from the cell surface to the endoplasmic reticulum (ER) of an intoxicated cell. The catalytic CtxA1 subunit then dissociates from the rest of the toxin, unfolds, and activates the ER-associated degradation system for export to the cytosol. Translocation occurs through an unusual ratchet mechanism in which the cytosolic chaperone Hsp90 couples CtxA1 refolding with CtxA1 extraction from the ER. Here, we report that Hsp90 recognises two peptide sequences from CtxA1: an N-terminal RPPDEI sequence (residues 11-16) and an LDIAPA sequence in the C-terminal region (residues 153-158) of the 192 amino acid protein. Peptides containing either sequence effectively blocked Hsp90 binding to full-length CtxA1. Both sequences were necessary for the ER-to-cytosol export of CtxA1. Mutagenesis studies further demonstrated that the RPP residues in the RPPDEI motif are required for CtxA1 translocation to the cytosol. The LDIAPA sequence is unique to CtxA1, but we identified an RPPDEI-like motif at the N- or C-termini of the A chains from four other ER-translocating toxins that act as ADP-ribosyltransferases: pertussis toxin, Escherichia coli heat-labile toxin, Pseudomonas aeruginosa exotoxin A, and Salmonella enterica serovar Typhimurium ADP-ribosylating toxin. Hsp90 plays a functional role in the intoxication process for most, if not all, of these toxins. Our work has established a defined RPPDEI binding motif for Hsp90 that is required for the ER-to-cytosol export of CtxA1 and possibly other toxin A chains as well.


Asunto(s)
Toxina del Cólera/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/metabolismo , Secuencias de Aminoácidos/genética , Animales , Toxinas Bacterianas/genética , Células CHO , Toxina del Cólera/química , Toxina del Cólera/genética , Toxina del Cólera/aislamiento & purificación , Cricetulus , Enterotoxinas/genética , Proteínas de Escherichia coli/genética , Exotoxinas/genética , Expresión Génica , Mutagénesis , Toxina del Pertussis/genética , Unión Proteica , Transporte de Proteínas/genética , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA