Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38558991

RESUMEN

The bacterial flagellum is an organelle utilized by many Gram-negative bacteria to facilitate motility. The flagellum is composed of a several µm long, extracellular filament that is connected to a cytoplasmic rotor-stator complex via a periplasmic rod. Composed of ∼20 structural proteins, ranging from a few subunits to several thousand building blocks, the flagellum is a paradigm of a complex macromolecular structure that utilizes a highly regulated assembly process. This process is governed by multiple checkpoints that ensure an ordered gene expression pattern coupled to the assembly of the various flagellar building blocks in order to produce a functional flagellum. Using epifluorescence, super-resolution STED and transmission electron microscopy, we discovered that in Salmonella , the absence of one periplasmic protein, FlhE, prevents proper flagellar morphogenesis and results in the formation of periplasmic flagella. The periplasmic flagella disrupt cell wall synthesis, leading to a loss of the standard cell morphology resulting in cell lysis. We propose a model where FlhE functions as a periplasmic chaperone to control assembly of the periplasmic rod to prevent formation of periplasmic flagella. Our results highlight that bacteria evolved sophisticated regulatory mechanisms to control proper flagellar assembly and minor deviations from this highly regulated process can cause dramatic physiological consequences.

2.
Development ; 149(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36093870

RESUMEN

Proven roles for hemocytes (blood cells) have expanded beyond the control of infections in Drosophila. Despite this, the crucial role of hemocytes in post-embryonic development has long thought to be limited to control of microorganisms during metamorphosis. This has previously been shown by rescue of adult development in hemocyte-ablation models under germ-free conditions. Here, we show that hemocytes have an essential role in post-embryonic development beyond their ability to control the microbiota. Using a newly generated strong hemocyte-specific driver line for the GAL4/UAS system, we show that specific ablation of hemocytes is early pupal lethal, even under axenic conditions. Genetic rescue experiments prove that this is a hemocyte-specific phenomenon. RNA-seq data suggests that dysregulation of the midgut is a prominent consequence of hemocyte ablation in larval stages, resulting in reduced gut lengths. Dissection suggests that multiple processes may be affected during metamorphosis. We believe this previously unreported role for hemocytes during metamorphosis is a major finding for the field.


Asunto(s)
Proteínas de Drosophila , Microbiota , Animales , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Desarrollo Embrionario , Hemocitos , Larva
3.
Nat Commun ; 13(1): 1030, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210413

RESUMEN

Coinfections with pathogenic microbes continually confront cervical mucosa, yet their implications in pathogenesis remain unclear. Lack of in-vitro models recapitulating cervical epithelium has been a bottleneck to study coinfections. Using patient-derived ectocervical organoids, we systematically modeled individual and coinfection dynamics of Human papillomavirus (HPV)16 E6E7 and Chlamydia, associated with carcinogenesis. The ectocervical stem cells were genetically manipulated to introduce E6E7 oncogenes to mimic HPV16 integration. Organoids from these stem cells develop the characteristics of precancerous lesions while retaining the self-renewal capacity and organize into mature stratified epithelium similar to healthy organoids. HPV16 E6E7 interferes with Chlamydia development and induces persistence. Unique transcriptional and post-translational responses induced by Chlamydia and HPV lead to distinct reprogramming of host cell processes. Strikingly, Chlamydia impedes HPV-induced mechanisms that maintain cellular and genome integrity, including mismatch repair in the stem cells. Together, our study employing organoids demonstrates the hazard of multiple infections and the unique cellular microenvironment they create, potentially contributing to neoplastic progression.


Asunto(s)
Chlamydia , Coinfección , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Reprogramación Celular/genética , Femenino , Papillomavirus Humano 16/genética , Humanos , Organoides , Microambiente Tumoral , Neoplasias del Cuello Uterino/genética
4.
Nat Commun ; 12(1): 6193, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702812

RESUMEN

Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.


Asunto(s)
Membrana Celular/metabolismo , Pared Celular/metabolismo , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Citotoxinas/metabolismo , Citotoxinas/toxicidad , Humanos , Leucocidinas/metabolismo , Leucocidinas/toxicidad , Lipopolisacáridos/genética , Lipopolisacáridos/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones , Fagocitos/efectos de los fármacos , Fosfatidilgliceroles/genética , Fosfatidilgliceroles/metabolismo , Transporte de Proteínas , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Ácidos Teicoicos/genética , Ácidos Teicoicos/metabolismo , Factores de Virulencia/toxicidad
5.
Gastroenterology ; 161(2): 623-636.e16, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33957136

RESUMEN

BACKGROUND & AIMS: The homeostasis of the gastrointestinal epithelium relies on cell regeneration and differentiation into distinct lineages organized inside glands and crypts. Regeneration depends on Wnt/ß-catenin pathway activation, but to understand homeostasis and its dysregulation in disease, we need to identify the signaling microenvironment governing cell differentiation. By using gastric glands as a model, we have identified the signals inducing differentiation of surface mucus-, zymogen-, and gastric acid-producing cells. METHODS: We generated mucosoid cultures from the human stomach and exposed them to different growth factors to obtain cells with features of differentiated foveolar, chief, and parietal cells. We localized the source of the growth factors in the tissue of origin. RESULTS: We show that epidermal growth factor is the major fate determinant distinguishing the surface and inner part of human gastric glands. In combination with bone morphogenetic factor/Noggin signals, epidermal growth factor controls the differentiation of foveolar cells vs parietal or chief cells. We also show that epidermal growth factor is likely to underlie alteration of the gastric mucosa in the precancerous condition atrophic gastritis. CONCLUSIONS: Use of our recently established mucosoid cultures in combination with analysis of the tissue of origin provided a robust strategy to understand differentiation and patterning of human tissue and allowed us to draw a new, detailed map of the signaling microenvironment in the human gastric glands.


Asunto(s)
Tipificación del Cuerpo/efectos de los fármacos , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Proteínas Portadoras/farmacología , Linaje de la Célula , Células Cultivadas , Microambiente Celular , Células Principales Gástricas/efectos de los fármacos , Células Principales Gástricas/metabolismo , Células Principales Gástricas/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestructura , Gastritis Atrófica/metabolismo , Gastritis Atrófica/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Organoides , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/ultraestructura , Vía de Señalización Wnt
6.
Viruses ; 13(4)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33918348

RESUMEN

Bacteriophages exert strong evolutionary pressure on their microbial hosts. In their lytic lifecycle, complete bacterial subpopulations are utilized as hosts for bacteriophage replication. However, during their lysogenic lifecycle, bacteriophages can integrate into the host chromosome and alter the host's genomic make-up, possibly resulting in evolutionary important adjustments. Not surprisingly, bacteria have evolved sophisticated immune systems to protect against phage infection. Streptococcus pyogenes isolates are frequently lysogenic and their prophages have been shown to be major contributors to the virulence of this pathogen. Most S. pyogenes phage research has focused on genomic prophages in relation to virulence, but little is known about the defensive arsenal of S. pyogenes against lytic phage infection. Here, we characterized Phage A1, an S. pyogenes bacteriophage, and investigated several mechanisms that S. pyogenes utilizes to protect itself against phage predation. We show that Phage A1 belongs to the Siphoviridae family and contains a circular double-stranded DNA genome that follows a modular organization described for other streptococcal phages. After infection, the Phage A1 genome can be detected in isolated S. pyogenes survivor strains, which enables the survival of the bacterial host and Phage A1 resistance. Furthermore, we demonstrate that the type II-A CRISPR-Cas system of S. pyogenes acquires new spacers upon phage infection, which are increasingly detectable in the absence of a capsule. Lastly, we show that S. pyogenes produces membrane vesicles that bind to phages, thereby limiting the pool of phages available for infection. Altogether, this work provides novel insight into survival strategies employed by S. pyogenes to combat phage predation.


Asunto(s)
Viabilidad Microbiana , Fagos de Streptococcus/genética , Fagos de Streptococcus/patogenicidad , Streptococcus pyogenes/fisiología , Streptococcus pyogenes/virología , Sistemas CRISPR-Cas , Genoma Viral , Lisogenia , Profagos/genética , Virulencia
7.
Sci Signal ; 14(673)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33688080

RESUMEN

Neutrophil extracellular traps (NETs) are structures consisting of chromatin and antimicrobial molecules that are released by neutrophils during a form of regulated cell death called NETosis. NETs trap invading pathogens, promote coagulation, and activate myeloid cells to produce type I interferons (IFNs), proinflammatory cytokines that regulate the immune system. Here, we showed that macrophages and other myeloid cells phagocytosed NETs. Once in phagosomes, NETs translocated to the cytosol, where the DNA backbones of these structures activated the innate immune sensor cyclic GMP-AMP synthase (cGAS) and induced type I IFN production. The NET-associated serine protease neutrophil elastase (NE) mediated the activation of this pathway. We showed that NET induction in mice treated with the lectin concanavalin A, a model of autoimmune hepatitis, resulted in cGAS-dependent stimulation of an IFN response, suggesting that NETs activated cGAS in vivo. Thus, our findings suggest that cGAS is a sensor of NETs, mediating immune cell activation during infection.


Asunto(s)
Trampas Extracelulares , Animales , Citosol , ADN , Ratones , Neutrófilos , Nucleotidiltransferasas/genética
8.
EMBO Mol Med ; 13(4): e13191, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33544398

RESUMEN

SARS-CoV-2, the agent that causes COVID-19, invades epithelial cells, including those of the respiratory and gastrointestinal mucosa, using angiotensin-converting enzyme-2 (ACE2) as a receptor. Subsequent inflammation can promote rapid virus clearance, but severe cases of COVID-19 are characterized by an inefficient immune response that fails to clear the infection. Using primary epithelial organoids from human colon, we explored how the central antiviral mediator IFN-γ, which is elevated in COVID-19, affects epithelial cell differentiation, ACE2 expression, and susceptibility to infection with SARS-CoV-2. In mouse and human colon, ACE2 is mainly expressed by surface enterocytes. Inducing enterocyte differentiation in organoid culture resulted in increased ACE2 production. IFN-γ treatment promoted differentiation into mature KRT20+ enterocytes expressing high levels of ACE2, increased susceptibility to SARS-CoV-2 infection, and resulted in enhanced virus production in infected cells. Similarly, infection-induced epithelial interferon signaling promoted enterocyte maturation and enhanced ACE2 expression. We here reveal a mechanism by which IFN-γ-driven inflammatory responses induce a vulnerable epithelial state with robust replication of SARS-CoV-2, which may have an impact on disease outcome and virus transmission.


Asunto(s)
COVID-19/etiología , Interferón gamma/inmunología , Modelos Inmunológicos , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/inmunología , COVID-19/patología , Diferenciación Celular/inmunología , Colon/inmunología , Colon/patología , Colon/virología , Susceptibilidad a Enfermedades , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/virología , Expresión Génica , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón gamma/administración & dosificación , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Ratones , Organoides/inmunología , Organoides/patología , Organoides/virología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Replicación Viral/inmunología
9.
mBio ; 11(5)2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32963006

RESUMEN

Carcinoma of the gallbladder (GBC) is the most frequent tumor of the biliary tract. Despite epidemiological studies showing a correlation between chronic infection with Salmonella enterica Typhi/Paratyphi A and GBC, the underlying molecular mechanisms of this fatal connection are still uncertain. The murine serovar Salmonella Typhimurium has been shown to promote transformation of genetically predisposed cells by driving mitogenic signaling. However, insights from this strain remain limited as it lacks the typhoid toxin produced by the human serovars Typhi and Paratyphi A. In particular, the CdtB subunit of the typhoid toxin directly induces DNA breaks in host cells, likely promoting transformation. To assess the underlying principles of transformation, we used gallbladder organoids as an infection model for Salmonella Paratyphi A. In this model, bacteria can invade epithelial cells, and we observed host cell DNA damage. The induction of DNA double-strand breaks after infection depended on the typhoid toxin CdtB subunit and extended to neighboring, non-infected cells. By cultivating the organoid derived cells into polarized monolayers in air-liquid interphase, we could extend the duration of the infection, and we observed an initial arrest of the cell cycle that does not depend on the typhoid toxin. Non-infected intoxicated cells instead continued to proliferate despite the DNA damage. Our study highlights the importance of the typhoid toxin in causing genomic instability and corroborates the epidemiological link between Salmonella infection and GBC.IMPORTANCE Bacterial infections are increasingly being recognized as risk factors for the development of adenocarcinomas. The strong epidemiological evidence linking Helicobacter pylori infection to stomach cancer has paved the way to the demonstration that bacterial infections cause DNA damage in the host cells, initiating transformation. In this regard, the role of bacterial genotoxins has become more relevant. Salmonella enterica serovars Typhi and Paratyphi A have been clinically associated with gallbladder cancer. By harnessing the stem cell potential of cells from healthy human gallbladder explant, we regenerated and propagated the epithelium of this organ in vitro and used these cultures to model S. Paratyphi A infection. This study demonstrates the importance of the typhoid toxin, encoded only by these specific serovars, in causing genomic instability in healthy gallbladder cells, posing intoxicated cells at risk of malignant transformation.


Asunto(s)
Daño del ADN , Células Epiteliales/microbiología , Células Epiteliales/patología , Vesícula Biliar/citología , Salmonella paratyphi A/patogenicidad , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Vesícula Biliar/microbiología , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Serogrupo , Virulencia/genética
10.
Mol Biochem Parasitol ; 235: 111247, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31874192

RESUMEN

Gliding motility and cell invasion are essential for the successful transmission of Plasmodium parasites. These processes rely on an acto-myosin motor located underneath the parasite plasma membrane. The Myosin A-tail interacting protein (MTIP) connects the class XIV myosin A (MyoA) to the gliding-associated proteins and is essential for assembly of the motor at the inner membrane complex. Here, we assessed the subcellular localization of MTIP in Plasmodium berghei motile stages from wild-type parasites and mutants that lack MyoA or the small heat shock protein 20 (HSP20). We demonstrate that MTIP is recruited to the apical end of motile ookinetes independently of the presence of MyoA. We also show that infective sporozoites displayed a polarized MTIP distribution during gliding, and that this distribution was abrogated in mutant parasites with an aberrant locomotion.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Locomoción/fisiología , Plasmodium berghei/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Proteínas de Choque Térmico/metabolismo , Proteínas de la Membrana/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo
11.
Gut ; 68(3): 400-413, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29467166

RESUMEN

OBJECTIVE: Helicobacter pylori causes life-long colonisation of the gastric mucosa, leading to chronic inflammation with increased risk of gastric cancer. Research on the pathogenesis of this infection would strongly benefit from an authentic human in vitro model. DESIGN: Antrum-derived gastric glands from surgery specimens served to establish polarised epithelial monolayers via a transient air-liquid interface culture stage to study cross-talk with H. pylori and the adjacent stroma. RESULTS: The resulting 'mucosoid cultures', so named because they recapitulate key characteristics of the gastric mucosa, represent normal stem cell-driven cultures that can be passaged for months. These highly polarised columnar epithelial layers encompass the various gastric antral cell types and secrete mucus at the apical surface. By default, they differentiate towards a foveolar, MUC5AC-producing phenotype, whereas Wnt signalling stimulates proliferation of MUC6-producing cells and preserves stemness-reminiscent of the gland base. Stromal cells from the lamina propria secrete Wnt inhibitors, antagonising stem-cell niche signalling and inducing differentiation. On infection with H. pylori, a strong inflammatory response is induced preferentially in the undifferentiated basal cell phenotype. Infection of cultures for several weeks produces foci of viable bacteria and a persistent inflammatory condition, while the secreted mucus establishes a barrier that only few bacteria manage to overcome. CONCLUSION: Gastric mucosoid cultures faithfully reproduce the features of normal human gastric epithelium, enabling new approaches for investigating the interaction of H. pylori with the epithelial surface and the cross-talk with the basolateral stromal compartment. Our observations provide striking insights in the regulatory circuits of inflammation and defence.


Asunto(s)
Mucosa Gástrica/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/fisiología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Infecciones por Helicobacter/metabolismo , Homeostasis/fisiología , Interacciones Microbiota-Huesped/fisiología , Humanos , Moco/metabolismo , Antro Pilórico/metabolismo , Antro Pilórico/microbiología , Antro Pilórico/patología , Nicho de Células Madre , Células del Estroma/fisiología , Técnicas de Cultivo de Tejidos/métodos
12.
Proc Natl Acad Sci U S A ; 115(32): E7568-E7577, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30038005

RESUMEN

Mosquito blood cells are immune cells that help control infection by vector-borne pathogens. Despite their importance, little is known about mosquito blood cell biology beyond morphological and functional criteria used for their classification. Here, we combined the power of single-cell RNA sequencing, high-content imaging flow cytometry, and single-molecule RNA hybridization to analyze a subset of blood cells of the malaria mosquito Anopheles gambiae By demonstrating that blood cells express nearly half of the mosquito transcriptome, our dataset represents an unprecedented view into their transcriptional program. Analyses of differentially expressed genes identified transcriptional signatures of two cell types and provide insights into the current classification of these cells. We further demonstrate the active transfer of a cellular marker between blood cells that may confound their identification. We propose that cell-to-cell exchange may contribute to cellular diversity and functional plasticity seen across biological systems.


Asunto(s)
Anopheles/genética , Células Sanguíneas/clasificación , Plasticidad de la Célula/genética , Malaria/transmisión , Mosquitos Vectores/genética , Animales , Animales Modificados Genéticamente , Anopheles/inmunología , Células Sanguíneas/inmunología , Comunicación Celular/genética , Conjuntos de Datos como Asunto , Femenino , Genómica/métodos , Mosquitos Vectores/inmunología , ARN/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma
13.
Immunity ; 49(1): 120-133.e9, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30005826

RESUMEN

B lymphocytes can suppress immunity through interleukin (IL)-10 production in infectious, autoimmune, and malignant diseases. Here, we have identified a natural plasma cell subset that distinctively expresses the inhibitory receptor LAG-3 and mediates this function in vivo. These plasma cells also express the inhibitory receptors CD200, PD-L1, and PD-L2. They develop from various B cell subsets in a B cell receptor (BCR)-dependent manner independently of microbiota in naive mice. After challenge they upregulate IL-10 expression via a Toll-like receptor-driven mechanism within hours and without proliferating. This function is associated with a unique transcriptome and epigenome, including the lowest amount of DNA methylation at the Il10 locus compared to other B cell subsets. Their augmented accumulation in naive mutant mice with increased BCR signaling correlates with the inhibition of memory T cell formation and vaccine efficacy after challenge. These natural regulatory plasma cells may be of broad relevance for disease intervention.


Asunto(s)
Antígenos CD/genética , Expresión Génica , Interleucina-10/biosíntesis , Células Plasmáticas/inmunología , Animales , Antígenos CD/inmunología , Subgrupos de Linfocitos B/inmunología , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Interleucina-10/genética , Activación de Linfocitos , Masculino , Ratones , Células Plasmáticas/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Salmonelosis Animal/inmunología , Transducción de Señal , Linfocitos T/inmunología , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/genética , Vacunas/inmunología , Proteína del Gen 3 de Activación de Linfocitos
14.
Cell Rep ; 23(3): 756-767, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29669282

RESUMEN

Proteins of the stomatin/prohibitin/flotillin/HfIK/C (SPFH) family are membrane-anchored and perform diverse cellular functions in different organelles. Here, we investigate the SPFH proteins of the murine malaria model parasite Plasmodium berghei, the conserved prohibitin 1, prohibitin 2, and stomatin-like protein and an unusual prohibitin-like protein (PHBL). The SPFH proteins localize to the parasite mitochondrion. While the conserved family members could not be deleted from the Plasmodium genome, PHBL was successfully ablated, resulting in impaired parasite fitness and attenuated virulence in the mammalian host. Strikingly, PHBL-deficient parasites fail to colonize the Anopheles vector because of complete arrest during ookinete development in vivo. We show that this arrest correlates with depolarization of the mitochondrial membrane potential (ΔΨmt). Our results underline the importance of SPFH proteins in the regulation of core mitochondrial functions and suggest that fine-tuning of ΔΨmt in malarial parasites is critical for colonization of the definitive host.


Asunto(s)
Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Represoras/metabolismo , Animales , Anopheles/parasitología , Interacciones Huésped-Parásitos , Insectos Vectores/parasitología , Estadios del Ciclo de Vida , Malaria/patología , Malaria/veterinaria , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/patogenicidad , Prohibitinas , Proteínas Protozoarias/genética , Proteínas Represoras/genética , Virulencia
15.
Autophagy ; 13(12): 2041-2055, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29251248

RESUMEN

The E3 ubiquitin ligase NEDD4 has been intensively studied in processes involved in viral infections, such as virus budding. However, little is known about its functions in bacterial infections. Our investigations into the role of NEDD4 in intracellular bacterial infections demonstrate that Mycobacterium tuberculosis and Listeria monocytogenes, but not Mycobacterium bovis BCG, replicate more efficiently in NEDD4 knockdown macrophages. In parallel, NEDD4 knockdown or knockout impaired basal macroautophagy/autophagy, as well as infection-induced autophagy. Conversely, NEDD4 expression promoted autophagy in an E3 catalytic activity-dependent manner, thereby restricting intracellular Listeria replication. Mechanistic studies uncovered that endogenous NEDD4 interacted with BECN1/Beclin 1 and this interaction increased during Listeria infection. Deficiency of NEDD4 resulted in elevated K48-linkage ubiquitination of endogenous BECN1. Further, NEDD4 mediated K6- and K27- linkage ubiquitination of BECN1, leading to elevated stability of BECN1 and increased autophagy. Thus, NEDD4 participates in killing of intracellular bacterial pathogens via autophagy by sustaining the stability of BECN1.


Asunto(s)
Autofagia , Bacterias/metabolismo , Membrana Celular/metabolismo , Espacio Intracelular/microbiología , Viabilidad Microbiana , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Beclina-1/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Fagosomas/metabolismo , Fagosomas/ultraestructura , Unión Proteica , Estabilidad Proteica , Células THP-1 , Ubiquitinación
16.
Chembiochem ; 18(10): 888-893, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28240414

RESUMEN

Neutrophils are short-lived leukocytes that migrate to sites of infection as part of the acute immune response, where they phagocytose, degranulate, and form neutrophil extracellular traps (NETs). During NET formation, the nuclear lobules of neutrophils disappear and the chromatin expands and, accessorized with neutrophilic granule proteins, is expelled. NETs can be pathogenic in, for example, sepsis, cancer, and autoimmune and cardiovascular diseases. Therefore, the identification of inhibitors of NET formation is of great interest. Screening of a focused library of natural-product-inspired compounds by using a previously validated phenotypic NET assay identified a group of tetrahydroisoquinolines as new NET formation inhibitors. This compound class opens up new avenues for the study of cellular death through NET formation (NETosis) at different stages, and might inspire new medicinal chemistry programs aimed at NET-dependent diseases.


Asunto(s)
Trampas Extracelulares/metabolismo , Lupus Eritematoso Sistémico/patología , Neutrófilos/metabolismo , Tetrahidroisoquinolinas/farmacología , Muerte Celular , Trampas Extracelulares/efectos de los fármacos , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos
17.
Front Immunol ; 7: 513, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27920776

RESUMEN

The pathogenic potential of neutrophil extracellular traps (NETs) was recently described, and their detection in tissue could serve as a prognostic marker. NETs are delicate and filigree structures; hence good tissue preservation is essential for their detection. Indeed, analysis of paraffin-embedded tissue has proven superior to the study of cryo sections. Though, under favorable conditions, the presence of NETs can be detected in tissue sections stained with histological dyes, definitive identification of NETs needs the colocalization of immunofluorescent signals for both nuclear and granular (or cytoplasmic) NET components. We tested diverse antigen retrieval methods and various combinations of commercially available antibodies and present here staining protocols to detect NETs in human and murine tissue sections.

18.
Mol Biol Cell ; 27(14): 2234-44, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226484

RESUMEN

Plasmodium relies on actin-based motility to migrate from the site of infection and invade target cells. Using a substrate-dependent gliding locomotion, sporozoites are able to move at fast speed (1-3 µm/s). This motility relies on a minimal set of actin regulatory proteins and occurs in the absence of detectable filamentous actin (F-actin). Here we report an overexpression strategy to investigate whether perturbations of F-actin steady-state levels affect gliding locomotion and host invasion. We selected two vital Plasmodium berghei G-actin-binding proteins, C-CAP and profilin, in combination with three stage-specific promoters and mapped the phenotypes afforded by overexpression in all three extracellular motile stages. We show that in merozoites and ookinetes, additional expression does not impair life cycle progression. In marked contrast, overexpression of C-CAP and profilin in sporozoites impairs circular gliding motility and salivary gland invasion. The propensity for productive motility correlates with actin accumulation at the parasite tip, as revealed by combinations of an actin-stabilizing drug and transgenic parasites. Strong expression of profilin, but not C-CAP, resulted in complete life cycle arrest. Comparative overexpression is an alternative experimental genetic strategy to study essential genes and reveals effects of regulatory imbalances that are not uncovered from deletion-mutant phenotyping.


Asunto(s)
Plasmodium/genética , Plasmodium/metabolismo , Profilinas/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Anopheles/parasitología , Movimiento Celular/genética , Movimiento Celular/fisiología , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Esporozoítos/fisiología
19.
Sci Rep ; 5: 12532, 2015 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-26219962

RESUMEN

The erythrocyte is an extraordinary host cell for intracellular pathogens and requires extensive remodelling to become permissive for infection. Malaria parasites modify their host red blood cells through protein export to acquire nutrients and evade immune responses. Endogenous fluorescent tagging of three signature proteins of the Plasmodium berghei translocon of exported proteins (PTEX), heat shock protein 101, exported protein 2 (EXP2), and PTEX88, revealed motile, tubular extensions of the parasitophorous vacuole that protrude from the parasite far into the red blood cell. EXP2 displays a more prominent presence at the periphery of the parasite, consistent with its proposed role in pore formation. The tubular compartment is most prominent during trophozoite growth. Distinct spatiotemporal expression of individual PTEX components during sporogony and liver-stage development indicates additional functions and tight regulation of the PTEX translocon during parasite life cycle progression. Together, live cell imaging and correlative light and electron microscopy permitted previously unrecognized spatiotemporal and subcellular resolution of PTEX-containing tubules in murine malaria parasites. These findings further refine current models for Plasmodium-induced erythrocyte makeover.


Asunto(s)
Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Estadios del Ciclo de Vida , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Transporte de Proteínas , Vacuolas/metabolismo , Vacuolas/ultraestructura
20.
Nature ; 512(7515): 387-92, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25119038

RESUMEN

The aryl hydrocarbon receptor (AhR) is a highly conserved ligand-dependent transcription factor that senses environmental toxins and endogenous ligands, thereby inducing detoxifying enzymes and modulating immune cell differentiation and responses. We hypothesized that AhR evolved to sense not only environmental pollutants but also microbial insults. We characterized bacterial pigmented virulence factors, namely the phenazines from Pseudomonas aeruginosa and the naphthoquinone phthiocol from Mycobacterium tuberculosis, as ligands of AhR. Upon ligand binding, AhR activation leads to virulence factor degradation and regulated cytokine and chemokine production. The relevance of AhR to host defence is underlined by heightened susceptibility of AhR-deficient mice to both P. aeruginosa and M. tuberculosis. Thus, we demonstrate that AhR senses distinct bacterial virulence factors and controls antibacterial responses, supporting a previously unidentified role for AhR as an intracellular pattern recognition receptor, and identify bacterial pigments as a new class of pathogen-associated molecular patterns.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Mycobacterium tuberculosis/inmunología , Pigmentos Biológicos/metabolismo , Pseudomonas aeruginosa/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Antibacterianos/metabolismo , Células de la Médula Ósea/citología , Citocinas/inmunología , Citocinas/metabolismo , Retroalimentación Fisiológica , Humanos , Ligandos , Activación de Macrófagos , Ratones , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/metabolismo , Fenazinas/metabolismo , Pigmentos Biológicos/química , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Piocianina/metabolismo , Factores de Virulencia/química , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA