Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(36): e2305649120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37639605

RESUMEN

Resilience to short-term perturbations, like inflammation, is a fundamental feature of microbiota, yet the underlying mechanisms of microbiota resilience are incompletely understood. Here, we show that Lactiplantibacillus plantarum, a major Drosophila commensal, stably colonizes the fruit fly gut during infection and is resistant to Drosophila antimicrobial peptides (AMPs). By transposon screening, we identified L. plantarum mutants sensitive to AMPs. These mutants were impaired in peptidoglycan O-acetylation or teichoic acid D-alanylation, resulting in increased negative cell surface charge and higher affinity to cationic AMPs. AMP-sensitive mutants were cleared from the gut after infection and aging-induced gut inflammation in wild-type, but not in AMP-deficient flies, suggesting that resistance to host AMPs is essential for commensal resilience in an inflamed gut environment. Thus, our work reveals that in addition to the host immune tolerance to the microbiota, commensal-encoded resilience mechanisms are necessary to maintain the stable association between host and microbiota during inflammation.


Asunto(s)
Péptidos Antimicrobianos , Drosophila , Animales , Péptidos Catiónicos Antimicrobianos/genética , Envejecimiento , Inflamación
2.
Int J Mol Sci ; 24(11)2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37298652

RESUMEN

Mouse guanylate-binding proteins (mGBPs) are recruited to various invasive pathogens, thereby conferring cell-autonomous immunity against these pathogens. However, whether and how human GBPs (hGBPs) target M. tuberculosis (Mtb) and L. monocytogenes (Lm) remains unclear. Here, we describe hGBPs association with intracellular Mtb and Lm, which was dependent on the ability of bacteria to induce disruption of phagosomal membranes. hGBP1 formed puncta structures which were recruited to ruptured endolysosomes. Furthermore, both GTP-binding and isoprenylation of hGBP1 were required for its puncta formation. hGBP1 was required for the recovery of endolysosomal integrity. In vitro lipid-binding assays demonstrated direct binding of hGBP1 to PI4P. Upon endolysosomal damage, hGBP1 was targeted to PI4P and PI(3,4)P2-positive endolysosomes in cells. Finally, live-cell imaging demonstrated that hGBP1 was recruited to damaged endolysosomes, and consequently mediated endolysosomal repair. In summary, we uncover a novel interferon-inducible mechanism in which hGBP1 contributes to the repair of damaged phagosomes/endolysosomes.


Asunto(s)
Proteínas de Unión al GTP , Fagosomas , Humanos , Animales , Ratones , Proteínas de Unión al GTP/metabolismo , Fagosomas/metabolismo , Interferones/metabolismo , Endosomas/metabolismo
3.
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
4.
Eur J Immunol ; 50(9): 1415, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33448355

RESUMEN

Retraction: Emoto, M., Emoto, Y., Yoshizawa, I., Kita, E., Shimizu, T., Hurwitz, R., Brinkmann, V. and Kaufmann, S.H.E. (2010), α-GalCer ameliorates listeriosis by accelerating infiltration of Gr-1+ cells into the liver. Eur. J. Immunol., 40: 1328-1341. DOI: https://doi.org/10.1002/eji.200939594 The above article, published online on 16 February 2010 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the Chairman of the Executive Committee of the European Journal of Immunology and Wiley-VCH Verlag GmbH & Co. KGaA. The retraction has been agreed following an investigation carried out by Gunma University (http://www.gunma-u.ac.jp/wp-content/uploads/2017/10/chosakekka29.pdf). The investigation was unable to determine the validity of the images for which Professor Emoto, the article's corresponding author, was responsible. As a result, the journal has made the decision to retract the article.

5.
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
6.
Eur J Immunol ; 49(4): 590-599, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30758851

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease that has high morbidity and can result in multi-organ damage. SLE is characterized by dysregulated activation of T- and B-lymphocytes and the production of autoantibodies directed against nuclear components. The endonuclease deoxyribonuclease 1 (DNase1) is abundant in blood and a subset of SLE patients have mutations in DNASE1. Furthermore, a report showed that Dnase1-deficient mice develop an SLE-like disease, but these mice also carry a deletion of the gene adjacent to Dnase1, which encodes the chaperone TRAP1/HSP75. We generated a murine strain deficient in Dnase1 with an intact Trap1 gene to examine if a lack of DNase1 is responsible for the development of a spontaneous SLE-like disease. We show that the Dnase1-deficient mice do indeed develop an SLE-like phenotype with elevated autoantibody production by 9 months and kidney damage by 12 months. Notably, this model recapitulates the female bias seen in human SLE patients since female Dnase1-deficient mice produced the highest concentrations of autoantibodies and had more severe kidney damage than males. Since there is currently no cure for SLE the protective role of DNase1 as demonstrated in our study remains of great therapeutic interest.


Asunto(s)
Desoxirribonucleasa I/deficiencia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/etiología , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Femenino , Estudios de Asociación Genética/métodos , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/etiología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Masculino , Ratones , Ratones Noqueados , Factores Sexuales
7.
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
8.
Mol Microbiol ; 110(5): 761-776, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30230643

RESUMEN

The human gastric pathogen Helicobacter pylori occurs in two basic variants, either exhibiting a functional cagPAI-encoded type-4-secretion-system (T4SS) or not. Only a few cagPAI-positive strains have been successfully adapted for long-term infection of mice, including the pre-mouse Sydney strain 1 (PMSS1). Here we confirm that PMSS1 induces gastric inflammation and neutrophil infiltration in mice, progressing to intestinal metaplasia. Complete genome analysis of PMSS1 revealed 1,423 coding sequences, encompassing the cagPAI gene cluster and, unusually, the location of the cytotoxin-associated gene A (cagA) approximately 15 kb downstream of the island. PMSS1 harbours three genetically exchangeable loci that are occupied by the hopQ coding sequences. HopQ represents a critical co-factor required for the translocation of CagA into the host cell and activation of NF-κB via the T4SS. Long-term colonisation of mice led to an impairment of cagPAI functionality. One of the bacterial clones re-isolated at four months post-infection revealed a mutation in the cagPAI gene cagW, resulting in a frame shift mutation, which prevented CagA translocation, possibly due to an impairment of T4SS function. Rescue of the mutant cagW re-established CagA translocation. Our data reveal intriguing insights into the adaptive abilities of PMSS1, suggesting functional modulation of the H. pylori cagPAI virulence attribute.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Sistemas de Secreción Tipo IV/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Familia de Multigenes , Virulencia , Secuenciación Completa del Genoma/métodos
9.
PLoS Pathog ; 13(10): e1006676, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29040326

RESUMEN

Mycobacterium tuberculosis (Mtb) primarily resides in the lung but can also persist in extrapulmonary sites. Macrophages are considered the prime cellular habitat in all tissues. Here we demonstrate that Mtb resides inside adipocytes of fat tissue where it expresses stress-related genes. Moreover, perigonadal fat of Mtb-infected mice disseminated the infection when transferred to uninfected animals. Adipose tissue harbors leukocytes in addition to adipocytes and other cell types and we observed that Mtb infection induces changes in adipose tissue biology depending on stage of infection. Mice infected via aerosol showed infiltration of inducible nitric oxide synthase (iNOS) or arginase 1 (Arg1)-negative F4/80+ cells, despite recruitment of CD3+, CD4+ and CD8+ T cells. Gene expression analysis of adipose tissue of aerosol Mtb-infected mice provided evidence for upregulated expression of genes associated with T cells and NK cells at 28 days post-infection. Strikingly, IFN-γ-producing NK cells and Mtb-specific CD8+ T cells were identified in perigonadal fat, specifically CD8+CD44-CD69+ and CD8+CD44-CD103+ subpopulations. Gene expression analysis of these cells revealed that they expressed IFN-γ and the lectin-like receptor Klrg1 and down-regulated CD27 and CD62L, consistent with an effector phenotype of Mtb-specific CD8+ T cells. Sorted NK cells expressed higher abundance of Klrg1 upon infection, as well. Our results reveal the ability of Mtb to persist in adipose tissue in a stressed state, and that NK cells and Mtb-specific CD8+ T cells infiltrate infected adipose tissue where they produce IFN-γ and assume an effector phenotype. We conclude that adipose tissue is a potential niche for Mtb and that due to infection CD8+ T cells and NK cells are attracted to this tissue.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/microbiología , Tuberculosis/inmunología , Tuberculosis/microbiología , Latencia del Virus/inmunología , Adipocitos/microbiología , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Células Asesinas Naturales/inmunología , Ratones , Mycobacterium tuberculosis/inmunología
10.
Nature ; 488(7413): 675-9, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22914092

RESUMEN

The blood­brain barrier (BBB) and the environment of the central nervous system (CNS) guard the nervous tissue from peripheral immune cells. In the autoimmune disease multiple sclerosis, myelin-reactive T-cell blasts are thought to transgress the BBB and create a pro-inflammatory environment in the CNS, thereby making possible a second autoimmune attack that starts from the leptomeningeal vessels and progresses into the parenchyma. Using a Lewis rat model of experimental autoimmune encephalomyelitis, we show here that contrary to the expectations of this concept, T-cell blasts do not efficiently enter the CNS and are not required to prepare the BBB for immune-cell recruitment. Instead, intravenously transferred T-cell blasts gain the capacity to enter the CNS after residing transiently within the lung tissues. Inside the lung tissues, they move along and within the airways to bronchus-associated lymphoid tissues and lung-draining mediastinal lymph nodes before they enter the blood circulation from where they reach the CNS. Effector T cells transferred directly into the airways showed a similar migratory pattern and retained their full pathogenicity. On their way the T cells fundamentally reprogrammed their gene-expression profile, characterized by downregulation of their activation program and upregulation of cellular locomotion molecules together with chemokine and adhesion receptors. The adhesion receptors include ninjurin 1, which participates in T-cell intravascular crawling on cerebral blood vessels. We detected that the lung constitutes a niche not only for activated T cells but also for resting myelin-reactive memory T cells. After local stimulation in the lung, these cells strongly proliferate and, after assuming migratory properties, enter the CNS and induce paralytic disease. The lung could therefore contribute to the activation of potentially autoaggressive T cells and their transition to a migratory mode as a prerequisite to entering their target tissues and inducing autoimmune disease.


Asunto(s)
Encéfalo/patología , Movimiento Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Pulmón/patología , Linfocitos T/patología , Traslado Adoptivo , Animales , Autoinmunidad/inmunología , Barrera Hematoencefálica/inmunología , Encéfalo/citología , Encéfalo/inmunología , Moléculas de Adhesión Celular Neuronal/metabolismo , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Memoria Inmunológica , Pulmón/citología , Pulmón/inmunología , Activación de Linfocitos , Vaina de Mielina/inmunología , Factores de Crecimiento Nervioso/metabolismo , Ratas , Ratas Endogámicas Lew , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
11.
EMBO J ; 32(14): 1977-89, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23736260

RESUMEN

We show that activation of Wnt/ß-catenin and attenuation of Bmp signals, by combined gain- and loss-of-function mutations of ß-catenin and Bmpr1a, respectively, results in rapidly growing, aggressive squamous cell carcinomas (SCC) in the salivary glands of mice. Tumours contain transplantable and hyperproliferative tumour propagating cells, which can be enriched by fluorescence activated cell sorting (FACS). Single mutations stimulate stem cells, but tumours are not formed. We show that ß-catenin, CBP and Mll promote self-renewal and H3K4 tri-methylation in tumour propagating cells. Blocking ß-catenin-CBP interaction with the small molecule ICG-001 and small-interfering RNAs against ß-catenin, CBP or Mll abrogate hyperproliferation and H3K4 tri-methylation, and induce differentiation of cultured tumour propagating cells into acini-like structures. ICG-001 decreases H3K4me3 at promoters of stem cell-associated genes in vitro and reduces tumour growth in vivo. Remarkably, high Wnt/ß-catenin and low Bmp signalling also characterize human salivary gland SCC and head and neck SCC in general. Our work defines mechanisms by which ß-catenin signals remodel chromatin and control induction and maintenance of tumour propagating cells. Further, it supports new strategies for the therapy of solid tumours.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína de Unión a CREB/antagonistas & inhibidores , Proteína de Unión a CREB/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Epigénesis Genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Histona Metiltransferasas , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Mutantes , Ratones SCID , Ratones Transgénicos , Mutación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pirimidinonas/farmacología , Neoplasias de las Glándulas Salivales/patología , Trasplante Heterólogo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores
12.
Traffic ; 15(4): 362-82, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24423236

RESUMEN

For membrane-bound intracellular pathogens, the surrounding vacuole is the portal of communication with the host cell. The parasitophorous vacuole (PV) harboring intrahepatocytic Plasmodium parasites satisfies the parasites' needs of nutrition and protection from host defenses to allow the rapid parasite growth that occurs during the liver stage of infection. In this study, we visualized the PV membrane (PVM) and the associated tubovesicular network (TVN) through fluorescent tagging of two PVM-resident Plasmodium berghei proteins, UIS4 and IBIS1. This strategy revealed previously unrecognized dynamics with which these membranes extend throughout the host cell. We observed dynamic vesicles, elongated clusters of membranes and long tubules that rapidly extend and contract from the PVM in a microtubule-dependent manner. Live microscopy, correlative light-electron microscopy and fluorescent recovery after photobleaching enabled a detailed characterization of these membranous features, including velocities, the distribution of UIS4 and IBIS1, and the connectivity of PVM and TVN. Labeling of host cell compartments revealed association of late endosomes and lysosomes with the elongated membrane clusters. Moreover, the signature host autophagosome protein LC3 was recruited to the PVM and TVN and colocalized with UIS4. Together, our data demonstrate that the membranes surrounding intrahepatic Plasmodium are involved in active remodeling of host cells.


Asunto(s)
Hígado/parasitología , Plasmodium/metabolismo , Animales , Membrana Celular/metabolismo , Interacciones Huésped-Parásitos , Plasmodium/patogenicidad
13.
Biochim Biophys Acta ; 1841(5): 745-58, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24239768

RESUMEN

Multiple Sclerosis (MS) is a chronic autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurodegeneration. Until recently, major therapeutic treatments have relied on agents requiring injection delivery. In September 2010, fingolimod/FTY720 (Gilenya, Novartis) was approved as the first oral treatment for relapsing forms of MS. Fingolimod causes down-modulation of S1P1 receptors on lymphocytes which prevents the invasion of autoaggressive T cells into the CNS. In astrocytes, down-modulation of S1P1 by the drug reduces astrogliosis, a hallmark of MS, thereby allowing restoration of productive astrocyte communication with other neural cells and the blood brain barrier. Animal data further suggest that the drug directly supports the recovery of nerve conduction and remyelination. In human MS, such mechanisms may explain the significant decrease in the number of inflammatory markers on brain magnetic resonance imaging in recent clinical trials, and the reduction of brain atrophy by the drug. Fingolimod binds to 4 of the 5 known S1P receptor subtypes, and significant efforts were made over the past 5 years to develop next generation S1P receptor modulators and determine the minimal receptor selectivity needed for maximal therapeutic efficacy in MS patients. Other approaches considered were competitive antagonists of the S1P1 receptor, inhibitors of the S1P lyase to prevent S1P degradation, and anti-S1P antibodies. Below we discuss the current status of the field, and the functional properties of the most advanced compounds. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.


Asunto(s)
Medicina Clínica , Inmunosupresores/farmacología , Lisofosfolípidos/metabolismo , Glicoles de Propileno/farmacología , Proyectos de Investigación , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Investigación Biomédica , Clorhidrato de Fingolimod , Humanos , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/metabolismo , Esfingosina/farmacología
14.
Infect Immun ; 83(9): 3648-56, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26150541

RESUMEN

Recently, we showed that endothelial heparan sulfate facilitates entry of a bacterial pathogen into the central nervous system. Here, we show that normal bactericidal activity of neutrophils is influenced by the sulfation pattern of heparan sulfate. Inactivation of heparan sulfate uronyl 2-O-sulfotransferase (Hs2st) in neutrophils substantially reduced their bactericidal activity, and Hs2st deficiency rendered mice more susceptible to systemic infection with the pathogenic bacterium group B Streptococcus. Specifically, altered sulfation of heparan sulfate in mutant neutrophils affected formation of neutrophil extracellular traps while not influencing phagocytosis, production of reactive oxygen species, or secretion of granular proteases. Heparan sulfate proteoglycan(s) is present in neutrophil extracellular traps, modulates histone affinity, and modulates their microbial activity. Hs2st-deficient brain endothelial cells show enhanced binding to group B Streptococcus and are more susceptible to apoptosis, likely contributing to the observed increase in dissemination of group B Streptococcus into the brain of Hs2st-deficient mice following intravenous challenge. Taken together, our data provide strong evidence that heparan sulfate from both neutrophils and the endothelium plays important roles in modulating innate immunity.


Asunto(s)
Células Endoteliales/inmunología , Proteoglicanos de Heparán Sulfato/inmunología , Inmunidad Innata/inmunología , Neutrófilos/inmunología , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Proteoglicanos de Heparán Sulfato/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Sulfotransferasas/metabolismo
15.
Mol Microbiol ; 94(6): 1285-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25315131

RESUMEN

Chlamydia trachomatis (Ctr), an obligate intracellular bacterium, survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to acquire nutrients. Ctr subverts cellular trafficking pathways from the Golgi by targeting small GTPases, including Rab proteins, to sustain intracellular bacterial replication; however, the precise mechanisms involved remain incompletely understood. Here, we show that Chlamydia infection in human epithelial cells induces microtubule remodeling, in particular the formation of detyrosinated stable MTs, to recruit Golgi ministacks, but not recycling endosomes, to the inclusion. These stable microtubules show increased resistance to chemically induced depolymerization, and their selective depletion results in reduced bacterial infectivity. Rab6 knockdown reversibly prevented not only Golgi ministack formation but also detyrosinated microtubule association with the inclusion. Our data demonstrate that Chlamydia co-opts the function of stable microtubules to support its development.


Asunto(s)
Infecciones por Chlamydia/patología , Chlamydia trachomatis/crecimiento & desarrollo , Aparato de Golgi/metabolismo , Microtúbulos/metabolismo , Infecciones por Chlamydia/metabolismo , Células HeLa , Células Hep G2 , Humanos , Microtúbulos/patología , Nocodazol/farmacología , Moduladores de Tubulina/farmacología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
16.
Eur J Immunol ; 44(8): 2380-93, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24782112

RESUMEN

General interest in the biological functions of IFN type I in Mycobacterium tuberculosis (Mtb) infection increased after the recent identification of a distinct IFN gene expression signature in tuberculosis (TB) patients. Here, we demonstrate that TB-susceptible mice lacking the receptor for IFN I (IFNAR1) were protected from death upon aerogenic infection with Mtb. Using this experimental model to mimic primary progressive pulmonary TB, we dissected the immune processes affected by IFN I. IFNAR1 signaling did not affect T-cell responses, but markedly altered migration of inflammatory monocytes and neutrophils to the lung. This process was orchestrated by IFNAR1 expressed on both immune and tissue-resident radioresistant cells. IFNAR1-driven TB susceptibility was initiated by augmented Mtb replication and in situ death events, along with CXCL5/CXCL1-driven accumulation of neutrophils in alveoli, followed by the discrete compartmentalization of Mtb in lung phagocytes. Early depletion of neutrophils rescued TB-susceptible mice to levels observed in mice lacking IFNAR1. We conclude that IFN I alters early innate events at the site of Mtb invasion leading to fatal immunopathology. These data furnish a mechanistic explanation for the detrimental role of IFN I in pulmonary TB and form a basis for understanding the complex roles of IFN I in chronic inflammation.


Asunto(s)
Interferón Tipo I/inmunología , Pulmón/inmunología , Fagocitos/inmunología , Transducción de Señal/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Células Cultivadas , Quimiocina CXCL1/inmunología , Quimiocina CXCL5/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Mycobacterium tuberculosis/inmunología , Neutrófilos/inmunología , Alveolos Pulmonares/inmunología , Receptor de Interferón alfa y beta/inmunología , Linfocitos T/inmunología
17.
Nature ; 457(7230): 731-5, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19060882

RESUMEN

The obligate intracellular bacterium Chlamydia trachomatis survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to obtain nutrients. Like many other intracellular pathogens, C. trachomatis has a marked requirement for host cell lipids, such as sphingolipids and cholesterol, produced in the endoplasmic reticulum and the Golgi apparatus. However, the mechanisms by which intracellular pathogens acquire host cell lipids are not well understood. In particular, no host cell protein responsible for transporting Golgi-derived lipids to the chlamydial inclusions has yet been identified. Here we show that Chlamydia infection in human epithelial cells induces Golgi fragmentation to generate Golgi ministacks surrounding the bacterial inclusion. Ministack formation is triggered by the proteolytic cleavage of the Golgi matrix protein golgin-84. Inhibition of golgin-84 truncation prevents Golgi fragmentation, causing a block in lipid acquisition and maturation of C. trachomatis. Golgi fragmentation by means of RNA-interference-mediated knockdown of distinct Golgi matrix proteins before infection enhances bacterial maturation. Our data functionally connect bacteria-induced golgin-84 cleavage, Golgi ministack formation, lipid acquisition and intracellular pathogen growth. We show that C. trachomatis subverts the structure and function of an entire host cell organelle for its own advantage.


Asunto(s)
Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/patogenicidad , Aparato de Golgi/microbiología , Aparato de Golgi/patología , Chlamydia muridarum/crecimiento & desarrollo , Células Epiteliales/microbiología , Células Epiteliales/patología , Técnicas de Silenciamiento del Gen , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi , Células HeLa , Humanos , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN , Proteínas de Transporte Vesicular
18.
Am J Respir Crit Care Med ; 190(9): 1053-66, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25275852

RESUMEN

RATIONALE: Myeloid cells encompass distinct populations with unique functions during homeostasis and disease. Recently, a novel subset of innate cells, myeloid-derived suppressor cells (MDSCs), has been described in cancer, which suppresses T-cell responses and fosters disease progression. The role of MDSCs in infection is insufficiently addressed. OBJECTIVES: To examine the presence and function of MDSCs during experimental pulmonary tuberculosis (TB) and further understand the immunologic consequences of direct interactions between MDSCs and lung bacterial pathogens. METHODS: Using cell-based approaches and experimental mouse models for pulmonary TB we characterized MDSCs as novel myeloid populations directly interacting with Mycobacterium tuberculosis (Mtb). MEASUREMENTS AND MAIN RESULTS: MDSCs readily phagocytosed Mtb, and released proinflammatory (IL-6, IL-1α) and immunomodulatory (IL-10) cytokines while retaining their suppressive capacity. MDSCs were identified at the site of infection in the lung in disease-resistant and -susceptible mice during pulmonary TB. Excessive MDSC accumulation in lungs correlated with elevated surface expression of IL-4Rα and heightened TB lethality, whereas targeted depletion of MDSCs ameliorated disease. CONCLUSIONS: Our data reveal that MDSCs provide a niche for pathogen survival and tailor immunity in TB. These findings suggest MDSCs as amenable targets for host-directed therapies and emphasize them as cellular-immune regulators during chronic inflammatory conditions, including chronic infections and microbial complications of neoplastic disorders.


Asunto(s)
Células Mieloides/fisiología , Tuberculosis Pulmonar/inmunología , Animales , Ratones , Ratones Endogámicos C57BL
19.
J Orthop Traumatol ; 16(3): 237-43, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25666724

RESUMEN

BACKGROUND: The aim of this prospective randomized study was to analyze migration and strain transmission of the Metha™ and Nanos™ femoral prostheses. MATERIALS AND METHODS: Between 1 January 2011 and 2 April 2013, 50 patients were randomized to receive short-stemmed femoral prostheses. Metha™ stems were implanted in 24 patients (12 female, 12 male; mean age 58.7 years; mean body mass index [BMI] 27.4) and Nanos™ stems in 26 patients (10 female, 16 male; mean age 59.7 years; mean BMI 27.1). Longitudinal stem migration, varus-valgus alignment, changes of center of rotation (COR), femoral offset and caput-collum-diaphyseal angle, leg length discrepancy, periprosthetic radiolucent lines incidence, and dual-energy X-ray absorptiometry (DEXA) scans were analysed after an average of 98 and 381 days. RESULTS: There was no significant change of varus-valgus alignment or clinically relevant migration of the Metha™ or Nanos™ prostheses during postoperative follow-up. After 12.3 months, the DEXA scans showed small but significant differences of bone mineral density in Gruen zones 1 (minus ~8 %) and 6 (plus ~9 %) for the Metha™ and in Gruen zone 1 (minus ~14 %) for the Nanos™ (paired t test). Visual analog scale (VAS) and Harris Hip Score (HHS) improved significantly for both implants (Nanos™/Metha™ 12.3 months postoperatively HSS 96.5/96.2; VAS 0.7/0.8, respectively). COR or offset did not change significantly after surgery. CONCLUSIONS: Neither implant showed signs of impaired osseointegration. DEXA demonstrated proximally located load transfer with only moderate proximal stress shielding. LEVEL OF EVIDENCE: II.


Asunto(s)
Artroplastia de Reemplazo de Cadera , Remodelación Ósea , Prótesis de Cadera , Osteoartritis de la Cadera/diagnóstico por imagen , Osteoartritis de la Cadera/cirugía , Falla de Prótesis , Absorciometría de Fotón , Adulto , Anciano , Densidad Ósea , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Diseño de Prótesis
20.
Am Heart J ; 168(5): 632-44, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25440790

RESUMEN

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) modulator, was the first oral disease-modifying therapy approved for relapsing forms of multiple sclerosis; it reduces autoreactive lymphocytes' egress from lymphoid tissues by down-regulating S1PRs. Sphingosine-1-phosphate signaling is implicated in a range of physiologic functions, and S1PRs are expressed differentially in various tissues, including the cardiovascular system. Modulation of S1PRs on cardiac cells provides an explanation for the transient effects of fingolimod on heart rate and atrioventricular conduction at initiation of fingolimod therapy, and for the mild but more persistent effects on blood pressure observed in some patients on long-term treatment. This review describes the nontherapeutic actions of fingolimod in the context of sphingosine-1-phosphate signaling in the cardiovascular system, as well as providing a summary of the associated clinical implications useful to physicians considering initiation of fingolimod therapy in patients. A transient reduction in heart rate (mean decrease of 8 beats per minute) and, less commonly, a temporary delay in atrioventricular conduction observed in some patients when initiating fingolimod therapy are both due to activation of S1PR subtype 1 on cardiac myocytes. These effects are a reflection of fingolimod first acting as a full S1PR agonist and thereafter functioning as an S1PR antagonist after down-regulation of S1PR subtype 1 at the cell surface. For most individuals, first-dose effects of fingolimod are asymptomatic, but all patients need to be monitored for at least 6 hours after the first dose, in accordance with the label recommendations.


Asunto(s)
Nodo Atrioventricular/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Inmunosupresores/farmacología , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/efectos de los fármacos , Esfingosina/análogos & derivados , Sistema Cardiovascular/efectos de los fármacos , Regulación hacia Abajo , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Esfingosina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA