Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
2.
Semin Cell Dev Biol ; 132: 230-241, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35177348

RESUMEN

Legionella pneumophila, a Gram-negative intracellular bacterium, is one of the major causes of Legionnaires' disease, a specific type of atypical pneumonia. Despite intensive research efforts that elucidated many relevant structural, molecular and medical insights into Legionella's pathogenicity, Legionnaires' disease continues to present an ongoing public health concern. Legionella's virulence is based on its ability to simultaneously hijack multiple molecular pathways of the host cell to ensure its fast replication and dissemination. Legionella usurps the host ubiquitin system through multiple effector proteins, using the advantage of both conventional and unconventional (phosphoribosyl-linked) ubiquitination, thus providing optimal conditions for its replication. In this review, we summarize the current understanding of L. pneumophila from medical, biochemical and molecular perspectives. We describe the clinical disease presentation, its diagnostics and treatment, as well as host-pathogen interactions, with the emphasis on the ability of Legionella to target the host ubiquitin system upon infection. Furthermore, the interdisciplinary use of innovative technologies enables better insights into the pathogenesis of Legionnaires' disease and provides new opportunities for its treatment and prevention.


Asunto(s)
Legionella pneumophila , Enfermedad de los Legionarios , Ubiquitina , Humanos , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Ubiquitina/metabolismo
3.
J Biol Chem ; 297(6): 101340, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34695417

RESUMEN

Legionella pneumophila is a facultative intracellular pathogen that uses the Dot/Icm Type IV secretion system (T4SS) to translocate many effectors into its host and establish a safe, replicative lifestyle. The bacteria, once phagocytosed, reside in a vacuolar structure known as the Legionella-containing vacuole (LCV) within the host cells and rapidly subvert organelle trafficking events, block inflammatory responses, hijack the host ubiquitination system, and abolish apoptotic signaling. This arsenal of translocated effectors can manipulate the host factors in a multitude of different ways. These proteins also contribute to bacterial virulence by positively or negatively regulating the activity of one another. Such effector-effector interactions, direct and indirect, provide the delicate balance required to maintain cellular homeostasis while establishing itself within the host. This review summarizes the recent progress in our knowledge of the structure-function relationship and biochemical mechanisms of select effector pairs from Legionella that work in opposition to one another, while highlighting the diversity of biochemical means adopted by this intracellular pathogen to establish a replicative niche within host cells.


Asunto(s)
Interacciones Huésped-Patógeno , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Sistemas de Secreción Tipo IV/metabolismo , Ubiquitinación , Vacuolas/metabolismo , Vacuolas/microbiología , Vacuolas/patología
4.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33468581

RESUMEN

Thymosin beta-4 (Tß4) is an actin-sequestering peptide that plays important roles in regeneration and remodeling of injured tissues. However, its function in a naturally occurring pathogenic bacterial infection model has remained elusive. We adopted Tß4-overexpressing transgenic (Tg) mice to investigate the role of Tß4 in acute pulmonary infection and systemic sepsis caused by Legionella pneumophila Upon infection, Tß4-Tg mice demonstrated significantly lower bacterial loads in the lung, less hyaline membranes and necrotic abscess, with lower interstitial infiltration of neutrophils, CD4+, and CD8+ T cells. Bronchoalveolar lavage fluid of Tß4-Tg mice possessed higher bactericidal activity against exogenously added L. pneumophila, suggesting that constitutive expression of Tß4 could efficiently control L. pneumophila Furthermore, qPCR analysis of lung homogenates demonstrated significant reduction of interleukin 1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α), which primarily originate from lung macrophages, in Tß4-Tg mice after pulmonary infection. Upon L. pneumophila challenge of bone marrow-derived macrophages (BMDM) in vitro, secretion of IL-1ß and TNF-α proteins was also reduced in Tß4-Tg macrophages, without affecting their survival. The anti-inflammatory effects of BMDM in Tß4-Tg mice on each cytokine were affected when triggering with tlr2, tlr4, tlr5, or tlr9 ligands, suggesting that anti-inflammatory effects of Tß4 are likely mediated by the reduced activation of Toll-like receptors (TLR). Finally, Tß4-Tg mice in a systemic sepsis model were protected from L. pneumophila-induced lethality compared to wild-type controls. Therefore, Tß4 confers effective resistance against L. pneumophila via two pathways, a bactericidal and an anti-inflammatory pathway, which can be harnessed to treat acute pneumonia and septic conditions caused by L. pneumophila in humans.


Asunto(s)
Resistencia a la Enfermedad/genética , Expresión Génica Ectópica , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/microbiología , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Timosina/genética , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/genética , Humanos , Inmunohistoquímica , Inmunofenotipificación , Enfermedad de los Legionarios/patología , Ligandos , Masculino , Ratones , Ratones Transgénicos , Neumonía Bacteriana/patología , Sepsis/genética , Sepsis/microbiología , Sepsis/patología , Receptores Toll-Like/metabolismo
5.
Nat Commun ; 12(1): 460, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469029

RESUMEN

Legionella pneumophila infects eukaryotic cells by forming a replicative organelle - the Legionella containing vacuole. During this process, the bacterial protein DrrA/SidM is secreted and manipulates the activity and post-translational modification (PTM) states of the vesicular trafficking regulator Rab1. As a result, Rab1 is modified with an adenosine monophosphate (AMP), and this process is referred to as AMPylation. Here, we use a chemical approach to stabilise low-affinity Rab:DrrA complexes in a site-specific manner to gain insight into the molecular basis of the interaction between the Rab protein and the AMPylation domain of DrrA. The crystal structure of the Rab:DrrA complex reveals a previously unknown non-conventional Rab-binding site (NC-RBS). Biochemical characterisation demonstrates allosteric stimulation of the AMPylation activity of DrrA via Rab binding to the NC-RBS. We speculate that allosteric control of DrrA could in principle prevent random and potentially cytotoxic AMPylation in the host, thereby perhaps ensuring efficient infection by Legionella.


Asunto(s)
Adenosina Monofosfato/metabolismo , Proteínas Bacterianas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/patología , Proteínas de Unión al GTP rab1/metabolismo , Regulación Alostérica , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/ultraestructura , Sitios de Unión/genética , Cristalografía por Rayos X , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/aislamiento & purificación , Factores de Intercambio de Guanina Nucleótido/ultraestructura , Guanosina Trifosfato/metabolismo , Humanos , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/microbiología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Fagocitosis , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab1/aislamiento & purificación , Proteínas de Unión al GTP rab1/ultraestructura
6.
Am J Respir Cell Mol Biol ; 64(5): 536-546, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33233920

RESUMEN

TOLLIP (Toll-interacting protein) is an intracellular adaptor protein with diverse actions throughout the body. In a context- and cell type-specific manner, TOLLIP can function as an inhibitor of inflammation and endoplasmic-reticulum stress, an activator of autophagy, or a critical regulator of intracellular vacuole trafficking. The distinct functions of this protein have been linked to innate immune responses and lung epithelial-cell apoptosis. TOLLIP genetic variants have been associated with a variety of chronic lung diseases, including idiopathic pulmonary fibrosis, asthma, and primary graft dysfunction after lung transplantation, and with infections, such as tuberculosis, Legionella pneumonia, and respiratory viruses. TOLLIP exists in a delicate homeostatic balance, with both positive and negative effects on the trajectory of pulmonary diseases. This translational review summarizes the genetic and molecular associations that link TOLLIP to the development and progression of noninfectious and infectious pulmonary diseases. We highlight current limitations of in vitro and in vivo models in assessing the role of TOLLIP in these conditions, and we describe future approaches that will enable a more nuanced exploration of the role of TOLLIP in pulmonary conditions. There has been a surge in recent research evaluating the role of this protein in human diseases, but critical mechanistic pathways require further exploration. By understanding its biologic functions in disease-specific contexts, we will be able to determine whether TOLLIP can be therapeutically modulated to treat pulmonary diseases.


Asunto(s)
Asma/genética , Rechazo de Injerto/genética , Fibrosis Pulmonar Idiopática/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Animales , Asma/inmunología , Asma/patología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/patología , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/inmunología , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Trasplante de Pulmón , Ratones , MicroARNs/genética , MicroARNs/inmunología , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/patología , Infecciones por Respirovirus/virología , Transducción de Señal , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
7.
PLoS One ; 15(11): e0241756, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33147266

RESUMEN

Sit Bath Systems (SBSs) are the most common hygiene method for patients who are not self-sufficient. Therefore, the water quality of SBSs in the nosocomial environment plays a fundamental role in controlling infections for both patients and health-care workers. A long-term study on Legionella and Pseudomonas aeruginosa (P. aeruginosa) contamination was performed in SBSs (n = 20) of six Health Care Facilities (HCFs). A total of 254 water samples were analyzed following ISO procedures. The samples were positive for P. aeruginosa (46.85%) and Legionella (53.54%), respectively, both over the directive limits. Legionella isolates were identified as: Legionella pneumophila (L. pneumophila) serogroups 1, 3, and 6 and Legionella non-pneumophila species (L. anisa, L. londiniensis, L. rubrilucens, and L. nagelii). Moreover, the contamination found was studied with respect to median temperature measured (42 °C), from which two groups (A and B) could be distinguished. P. aeruginosa was found in both groups (100% of SBSs), while a higher percentage of Legionella positive samples was found in group A (75% of SBSs), compared to group B (50% of SBSs), showing how Legionella control could be carried out by using temperatures above 42 °C. An analysis of SBS water pipelines, maintenance, and disinfection treatments indicates SBSs as a new source of infection risk for both patients and health-care workers.


Asunto(s)
Instituciones de Salud , Legionella/aislamiento & purificación , Microbiología del Agua , Proteínas Bacterianas/genética , Infección Hospitalaria/microbiología , Infección Hospitalaria/patología , ADN Bacteriano/química , ADN Bacteriano/metabolismo , Humanos , Legionella/genética , Legionella pneumophila/genética , Legionella pneumophila/aislamiento & purificación , Legionelosis/microbiología , Legionelosis/patología , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , Factores de Riesgo , Serogrupo , Temperatura
8.
Proc Natl Acad Sci U S A ; 117(25): 14433-14443, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513747

RESUMEN

During infection, the bacterial pathogen Legionella pneumophila manipulates a variety of host cell signaling pathways, including the Hippo pathway which controls cell proliferation and differentiation in eukaryotes. Our previous studies revealed that L. pneumophila encodes the effector kinase LegK7 which phosphorylates MOB1A, a highly conserved scaffold protein of the Hippo pathway. Here, we show that MOB1A, in addition to being a substrate of LegK7, also functions as an allosteric activator of its kinase activity. A crystallographic analysis of the LegK7-MOB1A complex revealed that the N-terminal half of LegK7 is structurally similar to eukaryotic protein kinases, and that MOB1A directly binds to the LegK7 kinase domain. Substitution of interface residues critical for complex formation abrogated allosteric activation of LegK7 both in vitro and within cells and diminished MOB1A phosphorylation. Importantly, the N-terminal extension (NTE) of MOB1A not only regulated complex formation with LegK7 but also served as a docking site for downstream substrates such as the transcriptional coregulator YAP1. Deletion of the NTE from MOB1A or addition of NTE peptides as binding competitors attenuated YAP1 recruitment to and phosphorylation by LegK7. By providing mechanistic insight into the formation and regulation of the LegK7-MOB1A complex, our study unravels a sophisticated molecular mimicry strategy that is used by L. pneumophila to take control of the host cell Hippo pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Bacterianas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Legionella pneumophila/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Regulación Alostérica , Animales , Proteínas Bacterianas/genética , Proteínas de Ciclo Celular/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones , Simulación de Dinámica Molecular , Imitación Molecular , Fosforilación , Unión Proteica , Proteínas Quinasas/genética , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
9.
Annu Rev Pathol ; 15: 439-466, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31657966

RESUMEN

Legionella species are environmental gram-negative bacteria able to cause a severe form of pneumonia in humans known as Legionnaires' disease. Since the identification of Legionella pneumophila in 1977, four decades of research on Legionella biology and Legionnaires' disease have brought important insights into the biology of the bacteria and the molecular mechanisms that these intracellular pathogens use to cause disease in humans. Nowadays, Legionella species constitute a remarkable model of bacterial adaptation, with a genus genome shaped by their close coevolution with amoebae and an ability to exploit many hosts and signaling pathways through the secretion of a myriad of effector proteins, many of which have a eukaryotic origin. This review aims to discuss current knowledge of Legionella infection mechanisms and future research directions to be taken that might answer the many remaining open questions. This research will without a doubt be a terrific scientific journey worth taking.


Asunto(s)
Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Adaptación Fisiológica/genética , Adaptación Fisiológica/inmunología , Amoeba/genética , Amoeba/inmunología , Amoeba/patogenicidad , Células Eucariotas/inmunología , Células Eucariotas/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Legionella/clasificación , Legionella/genética , Legionella/inmunología , Legionella pneumophila/genética , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/patología
10.
Sci Rep ; 9(1): 13295, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31527638

RESUMEN

Neutrophil elastase is a serine protease that has been implicated in the pathogenesis of inflammatory bowel disease. Due to post-translational control of its activation and high expression of its inhibitors in the gut, measurements of total expression poorly reflect the pool of active, functional neutrophil elastase. Fluorogenic substrate probes have been used to measure neutrophil elastase activity, though these tools lack specificity and traceability. PK105 is a recently described fluorescent activity-based probe, which binds to neutrophil elastase in an activity-dependent manner. The irreversible nature of this probe allows for accurate identification of its targets in complex protein mixtures. We describe the reactivity profile of PK105b, a new analogue of PK105, against recombinant serine proteases and in tissue extracts from healthy mice and from models of inflammation induced by oral cancer and Legionella pneumophila infection. We apply PK105b to measure neutrophil elastase activation in an acute model of experimental colitis. Neutrophil elastase activity is detected in inflamed, but not healthy, colons. We corroborate this finding in mucosal biopsies from patients with ulcerative colitis. Thus, PK105b facilitates detection of neutrophil elastase activity in tissue lysates, and we have applied it to demonstrate that this protease is unequivocally activated during colitis.


Asunto(s)
Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Elastasa de Leucocito/metabolismo , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias de la Boca/patología
11.
Cell Host Microbe ; 26(4): 551-563.e6, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31540829

RESUMEN

During infection, Legionella pneumophila translocates over 300 effector proteins into the host cytosol, allowing the pathogen to establish an endoplasmic reticulum (ER)-like Legionella-containing vacuole (LCV) that supports bacterial replication. Here, we perform a genome-wide CRISPR-Cas9 screen and secondary targeted screens in U937 human monocyte/macrophage-like cells to systematically identify host factors that regulate killing by L. pneumophila. The screens reveal known host factors hijacked by L. pneumophila, as well as genes spanning diverse trafficking and signaling pathways previously not linked to L. pneumophila pathogenesis. We further characterize C1orf43 and KIAA1109 as regulators of phagocytosis and show that RAB10 and its chaperone RABIF are required for optimal L. pneumophila replication and ER recruitment to the LCV. Finally, we show that Rab10 protein is recruited to the LCV and ubiquitinated by the effectors SidC/SdcA. Collectively, our results provide a wealth of previously undescribed insights into L. pneumophila pathogenesis and mammalian cell function.


Asunto(s)
Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/patología , Fagocitosis/inmunología , Proteínas/genética , Vacuolas/microbiología , Animales , Proteínas Bacterianas/metabolismo , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Retículo Endoplásmico/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/genética , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Células RAW 264.7 , Células U937 , Factores de Virulencia/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
12.
Proc Natl Acad Sci U S A ; 116(36): 17775-17785, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31431530

RESUMEN

Legionella pneumophila causes a potentially fatal form of pneumonia by replicating within macrophages in the Legionella-containing vacuole (LCV). Bacterial survival and proliferation within the LCV rely on hundreds of secreted effector proteins comprising high functional redundancy. The vacuolar membrane-localized MavN, hypothesized to support iron transport, is unique among effectors because loss-of-function mutations result in severe intracellular growth defects. We show here an iron starvation response by L. pneumophila after infection of macrophages that was prematurely induced in the absence of MavN, consistent with MavN granting access to limiting cellular iron stores. MavN cysteine accessibilities to a membrane-impermeant label were determined during macrophage infections, revealing a topological pattern supporting multipass membrane transporter models. Mutations to several highly conserved residues that can take part in metal recognition and transport resulted in defective intracellular growth. Purified MavN and mutant derivatives were directly tested for transporter activity after heterologous purification and liposome reconstitution. Proteoliposomes harboring MavN exhibited robust transport of Fe2+, with the severity of defect of most mutants closely mimicking the magnitude of defects during intracellular growth. Surprisingly, MavN was equivalently proficient at transporting Fe2+, Mn2+, Co2+, or Zn2+ Consequently, flooding infected cells with either Mn2+ or Zn2+ allowed collaboration with iron to enhance intracellular growth of L. pneumophila ΔmavN strains, indicating a clear role for MavN in transporting each of these ions. These findings reveal that MavN is a transition-metal-ion transporter that plays a critical role in response to iron limitation during Legionella infection.


Asunto(s)
Proteínas Bacterianas , Proteínas de Transporte de Catión , Legionella pneumophila , Metales/metabolismo , Vacuolas , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/metabolismo , Enfermedad de los Legionarios/patología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Células U937 , Vacuolas/genética , Vacuolas/metabolismo
13.
J Infect Dis ; 220(10): 1700-1710, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31268152

RESUMEN

BACKGROUND: Legionella can cause Legionnaires' disease, a potentially fatal form of pneumonia that occurs as sporadic epidemics. Not all strains display the same propensity to cause disease in humans. Because Legionella pneumophila serogroup 1 is responsible for >85% of infections, the majority of studies have examined this serogroup, but there are 3 commonly used laboratory strains: L pneumophila serogroup 1 Philadelphia (Phil-1)-derived strains JR32 and Lp01 and 130b-derived strain AA100. METHODS: We evaluated the ability of Phil-1, JR32, Lp01, and AA100 to cause disease in guinea pigs. RESULTS: We found that, although Phil-1, JR32, and AA100 cause an acute pneumonia and death by 4 days postinfection (100%), strain Lp01 does not cause mortality (0%). We also noted that Lp01 lacks a mobile element, designated p45, whose presence correlates with virulence. Transfer of p45 into Lp01 results in recovery of the ability of this strain to cause mortality, leads to more pronounced disease, and correlates with increased interferon-γ levels in the lungs and spleens before death. CONCLUSIONS: These observations suggest a mechanism of Legionnaires' disease pathogenesis due to the presence of type IVA secretion systems that cause higher mortality due to overinduction of a proinflammatory response in the host.


Asunto(s)
Secuencias Repetitivas Esparcidas , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/patología , Enfermedad de los Legionarios/fisiopatología , Sistemas de Secreción Tipo IV/genética , Factores de Virulencia/genética , Animales , Modelos Animales de Enfermedad , Cobayas , Interferón gamma/análisis , Enfermedad de los Legionarios/inmunología , Pulmón/patología , Bazo/patología , Análisis de Supervivencia
14.
PLoS Pathog ; 15(6): e1007886, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31251782

RESUMEN

Inflammasomes are cytosolic multi-protein complexes that detect infection or cellular damage and activate the Caspase-1 (CASP1) protease. The NAIP5/NLRC4 inflammasome detects bacterial flagellin and is essential for resistance to the flagellated intracellular bacterium Legionella pneumophila. The effectors required downstream of NAIP5/NLRC4 to restrict bacterial replication remain unclear. Upon NAIP5/NLRC4 activation, CASP1 cleaves and activates the pore-forming protein Gasdermin-D (GSDMD) and the effector caspase-7 (CASP7). However, Casp1-/- (and Casp1/11-/-) mice are only partially susceptible to L. pneumophila and do not phenocopy Nlrc4-/-mice, because NAIP5/NLRC4 also activates CASP8 for restriction of L. pneumophila infection. Here we show that CASP8 promotes the activation of CASP7 and that Casp7/1/11-/- and Casp8/1/11-/- mice recapitulate the full susceptibility of Nlrc4-/- mice. Gsdmd-/- mice exhibit only mild susceptibility to L. pneumophila, but Gsdmd-/-Casp7-/- mice are as susceptible as the Nlrc4-/- mice. These results demonstrate that GSDMD and CASP7 are the key substrates downstream of NAIP5/NLRC4/CASP1/8 required for resistance to L. pneumophila.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , Caspasa 1/inmunología , Caspasa 7/inmunología , Caspasa 8/inmunología , Inflamasomas/inmunología , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Caspasa 1/genética , Caspasa 7/genética , Caspasa 8/genética , Inflamasomas/genética , Péptidos y Proteínas de Señalización Intracelular , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/patología , Ratones , Ratones Noqueados , Proteína Inhibidora de la Apoptosis Neuronal/genética , Proteínas de Unión a Fosfato
15.
J Biol Chem ; 294(16): 6405-6415, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30733336

RESUMEN

Upon phagocytosis into macrophages, the intracellular bacterial pathogen Legionella pneumophila secretes effector proteins that manipulate host cell components, enabling it to evade lysosomal degradation. However, the bacterial proteins involved in this evasion are incompletely characterized. Here we show that the L. pneumophila effector protein RavD targets host membrane compartments and contributes to the molecular mechanism the pathogen uses to prevent encounters with lysosomes. Protein-lipid binding assays revealed that RavD selectively binds phosphatidylinositol-3-phosphate (PI(3)P) in vitro We further determined that a C-terminal RavD region mediates the interaction with PI(3)P and that this interaction requires Arg-292. In transiently transfected mammalian cells, mCherry-RavD colocalized with the early endosome marker EGFP-Rab5 as well as the PI(3)P biosensor EGFP-2×FYVE. However, treatment with the phosphoinositide 3-kinase inhibitor wortmannin did not disrupt localization of mCherry-RavD to endosomal compartments, suggesting that RavD's interaction with PI(3)P is not necessary to anchor RavD to endosomal membranes. Using superresolution and immunogold transmission EM, we observed that, upon translocation into macrophages, RavD was retained onto the Legionella-containing vacuole and was also present on small vesicles adjacent to the vacuole. We also report that despite no detectable effects on intracellular growth of L. pneumophila within macrophages or amebae, the lack of RavD significantly increased the number of vacuoles that accumulate the late endosome/lysosome marker LAMP-1 during macrophage infection. Together, our findings suggest that, although not required for intracellular replication of L. pneumophila, RavD is a part of the molecular mechanism that steers the Legionella-containing vacuole away from endolysosomal maturation pathways.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endosomas/metabolismo , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/genética , Endosomas/genética , Endosomas/ultraestructura , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/patología , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/genética , Lisosomas/ultraestructura , Macrófagos/microbiología , Macrófagos/ultraestructura , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/antagonistas & inhibidores , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Células U937 , Vacuolas/genética , Vacuolas/microbiología , Vacuolas/ultraestructura , Wortmanina/farmacología , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
16.
Methods Mol Biol ; 1921: 323-331, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30694502

RESUMEN

Legionnaires' disease is a severe pneumonia caused by inhalation of Legionella pneumophila. Although powerful infection models ranging from monocellular host systems to mammals were developed, numerous intra- and extracellular interactions of L. pneumophila factors with human lung tissue structures remain unknown. Therefore, we developed and applied a novel infection model for Legionnaires' disease comprising living human lung tissue explants (HLTEs). This model allows analyzing Legionella infections at a unique level of complexity and narrows the gap between current infection models and postmortem histopathology analyses of infected patients. Here we describe the infection of tumor-free pulmonary tissue samples from patients undergoing lobe- or pneumectomy because of lung cancer. The method comprises bacterial cultivation, preparation of HLTEs, and infection of HLTEs. The infected tissue samples allow to characterize tissue damage, bacterial localization, dissemination and growth kinetics, and the host's molecular response.


Asunto(s)
Legionella pneumophila/fisiología , Enfermedad de los Legionarios/microbiología , Pulmón/microbiología , Interacciones Huésped-Patógeno , Humanos , Enfermedad de los Legionarios/patología , Pulmón/patología , Técnicas de Cultivo de Tejidos
17.
Methods Mol Biol ; 1921: 399-417, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30694506

RESUMEN

Infection of C57BL/6 mice with wild-type Legionella pneumophila typically results in very mild disease. However, in mice where the cytosolic recognition of flagellin is impaired by mutation, L. pneumophila infection results in more severe lung inflammation that is reminiscent of Legionnaires' disease. This can be replicated in wild-type mice by using aflagellated mutants of L. pneumophila. These models greatly facilitate the investigation of L. pneumophila virulence factors and the complex pulmonary immune system that is triggered by infection. Here we describe methods for infecting C57BL/6 mice with aflagellated L. pneumophila, the quantification of bacterial load in the lungs and isolation and analysis of invading immune cells. These assays enable the identification of phagocyte subsets and can determine whether phagocytic cells act as a replicative niche for L. pneumophila replication.


Asunto(s)
Interacciones Huésped-Patógeno , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/microbiología , Animales , Carga Bacteriana , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/patología , Pulmón/metabolismo , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología
18.
Med Microbiol Immunol ; 208(1): 25-32, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30386929

RESUMEN

Gram-negative bacterium Legionella is able to proliferate intracellularly in mammalian host cells and amoeba, which became known in 1976 since they caused a large outbreak of pneumonia. It had been reported that different strains of Legionella pneumophila, Legionella micdadei, Legionella longbeachae, and Legionella feeleii caused human respiratory diseases, which were known as Pontiac fever or Legionnaires' disease. However, the differences of the virulence traits among the strains of the single species and the pathogenesis of the two diseases that were due to the bacterial virulence factors had not been well elucidated. L. feeleii is an important pathogenic organism in Legionellae, which attracted attention due to cause an outbreak of Pontiac fever in 1981 in Canada. In published researches, it has been found that L. feeleii serogroup 2 (ATCC 35849, LfLD) possess mono-polar flagellum, and L. feeleii serogroup 1 (ATCC 35072, WRLf) could secrete some exopolysaccharide (EPS) materials to the surrounding. Although the virulence of the L. feeleii strain was evidenced that could be promoted, the EPS might be dispensable for the bacteria that caused Pontiac fever. Based on the current knowledge, we focused on bacterial infection in human and murine host cells, intracellular growth, cytopathogenicity, stimulatory capacity of cytokines secretion, and pathogenic effects of the EPS of L. feeleii in this review.


Asunto(s)
Citocinas/metabolismo , Legionella/patogenicidad , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Polisacáridos Bacterianos/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular , Humanos , Legionella/crecimiento & desarrollo , Ratones , Polisacáridos Bacterianos/toxicidad , Virulencia , Factores de Virulencia/toxicidad
19.
Biochem Biophys Res Commun ; 508(2): 608-613, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30509489

RESUMEN

BACKGROUND: Inhalation of aerosolized Legionella pneumophila, a Gram-negative bacterium, can cause severe pneumonia. During infection, L. pneumophila replicates intracellularly in macrophages. The involvement of host microRNAs (miRNAs) in L. pneumophila infection is not fully understood. METHODS: The human macrophage-like cell line U937 was infected with L. pneumophila. The levels of miRNA and messenger RNA (mRNA) were measured using reverse transcriptase polymerase chain reaction. Release of lactate dehydrogenase was used to evaluate cytotoxicity. The expression of RICTOR and related proteins was examined by western blotting of cell lysates. RESULTS: L. pneumophila infection upregulated the expression of miR-218 and the host genes SLIT2 and SLIT3 in U937 cells. The expression of RICTOR, a component of the mechanistic target of rapamycin complex 2 (mTORC2), decreased during L. pneumophila infection. RICTOR protein expression was inhibited by the overexpression of miR-218, whereas knockdown of miR-218 restored the downregulation of RICTOR by L. pneumophila. L. pneumophila infection induced the expression of the proinflammatory cytokines IL-6 and TNF-alpha, which was modulated by knockdown of miR-218 or RICTOR. CONCLUSIONS: Our study revealed the involvement of miR-218 in regulating the inflammatory response of macrophages against L. pneumophila infection. These findings suggest potential novel roles for miR-218 and RICTOR as therapeutic targets of L. pneumophila infection.


Asunto(s)
Legionella pneumophila , Enfermedad de los Legionarios/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Citocinas , Interacciones Huésped-Patógeno , Humanos , Inflamación , Enfermedad de los Legionarios/patología , Enfermedad de los Legionarios/virología , Macrófagos/microbiología , Macrófagos/patología , MicroARNs/análisis , ARN Mensajero/análisis , Células U937
20.
Infect Immun ; 87(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30323031

RESUMEN

Legionella pneumophila causes life-threatening pneumonia culminating in acute lung injury. Innate and adaptive cytokines play an important role in host defense against L. pneumophila infection. Interleukin-36 (IL-36) cytokines are recently described members of the larger IL-1 cytokine family known to exert potent inflammatory effects. In this study, we elucidated the role for IL-36 cytokines in experimental pneumonia caused by L. pneumophila Intratracheal (i.t.) administration of L. pneumophila induced the upregulation of both IL-36α and IL-36γ mRNA and protein production in the lung. Compared to the findings for L. pneumophila-infected wild-type (WT) mice, the i.t. administration of L. pneumophila to IL-36 receptor-deficient (IL-36R-/-) mice resulted in increased mortality, a delay in lung bacterial clearance, increased L. pneumophila dissemination to extrapulmonary organs, and impaired glucose homeostasis. Impaired lung bacterial clearance in IL-36R-/- mice was associated with a significantly reduced accumulation of inflammatory cells and the decreased production of proinflammatory cytokines and chemokines. Ex vivo, reduced expression of costimulatory molecules and impaired M1 polarization were observed in alveolar macrophages isolated from infected IL-36R-/- mice compared to macrophages from WT mice. While L. pneumophila-induced mortality in IL-36α- or IL-36γ-deficient mice was not different from that in WT animals, antibody-mediated neutralization of IL-36γ in IL-36α-/- mice resulted in mortality similar to that observed in IL-36R-/- mice, indicating redundant and overlapping roles for these cytokines in experimental murine L. pneumophila pneumonia.


Asunto(s)
Interleucina-1/metabolismo , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/patología , Animales , Modelos Animales de Enfermedad , Femenino , Interleucina-1/deficiencia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...