Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Clin Exp Immunol ; 218(1): 40-54, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-38280212

RESUMEN

The intertwined interactions various immune cells have with epithelial cells in our body require sophisticated experimental approaches to be studied. Due to the limitations of immortalized cell lines and animal models, there is an increasing demand for human in vitro model systems to investigate the microenvironment of immune cells in normal and in pathological conditions. Organoids, which are self-renewing, 3D cellular structures that are derived from stem cells, have started to provide gap-filling tissue modelling solutions. In this review, we first demonstrate with some of the available examples how organoid-based immune cell co-culture experiments can advance disease modelling of cancer, inflammatory bowel disease, and tissue regeneration. Then, we argue that to achieve both complexity and scale, organ-on-chip models combined with cutting-edge microfluidics-based technologies can provide more precise manipulation and readouts. Finally, we discuss how genome editing techniques and the use of patient-derived organoids and immune cells can improve disease modelling and facilitate precision medicine. To achieve maximum impact and efficiency, these efforts should be supported by novel infrastructures such as organoid biobanks, organoid facilities, as well as drug screening and host-microbe interaction testing platforms. All these together or in combination can allow researchers to shed more detailed, and often patient-specific, light on the crosstalk between immune cells and epithelial cells in health and disease.


Asunto(s)
Técnicas de Cocultivo , Organoides , Humanos , Organoides/inmunología , Técnicas de Cocultivo/métodos , Animales , Medicina de Precisión/métodos , Dispositivos Laboratorio en un Chip , Enfermedades Inflamatorias del Intestino/inmunología , Neoplasias/inmunología
2.
PLoS Comput Biol ; 17(2): e1008685, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33534793

RESUMEN

The SARS-CoV-2 pandemic of 2020 has mobilised scientists around the globe to research all aspects of the coronavirus virus and its infection. For fruitful and rapid investigation of viral pathomechanisms, a collaborative and interdisciplinary approach is required. Therefore, we have developed ViralLink: a systems biology workflow which reconstructs and analyses networks representing the effect of viruses on intracellular signalling. These networks trace the flow of signal from intracellular viral proteins through their human binding proteins and downstream signalling pathways, ending with transcription factors regulating genes differentially expressed upon viral exposure. In this way, the workflow provides a mechanistic insight from previously identified knowledge of virally infected cells. By default, the workflow is set up to analyse the intracellular effects of SARS-CoV-2, requiring only transcriptomics counts data as input from the user: thus, encouraging and enabling rapid multidisciplinary research. However, the wide-ranging applicability and modularity of the workflow facilitates customisation of viral context, a priori interactions and analysis methods. Through a case study of SARS-CoV-2 infected bronchial/tracheal epithelial cells, we evidence the functionality of the workflow and its ability to identify key pathways and proteins in the cellular response to infection. The application of ViralLink to different viral infections in a context specific manner using different available transcriptomics datasets will uncover key mechanisms in viral pathogenesis.


Asunto(s)
COVID-19/metabolismo , Biología Computacional/métodos , Regulación Viral de la Expresión Génica , SARS-CoV-2/patogenicidad , Transducción de Señal , Algoritmos , Bronquios/virología , Análisis por Conglomerados , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Investigación Interdisciplinaria , Pulmón/virología , Modelos Estadísticos , Biología de Sistemas , Transcriptoma , Flujo de Trabajo
3.
Glycobiology ; 25(1): 42-54, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25187161

RESUMEN

Intestinal γδ T-cell receptor-bearing intraepithelial lymphocytes (γδ IELs) play a multifaceted role in maintaining mucosal homeostasis. In order to investigate the relationship between O-glycosylation and inflammation, we carried out an in-depth mass spectrometric comparison of the intestinal O-glycosylation profile of mice lacking γδ IELs (TCRδ(-/-)) and of their wild-type (WT) littermates. A total of 69 nonsulfated and 59 sulfated compositional types of O-glycans were identified in the small intestine and colon of TCRδ(-/-) and WT mice. Our results demonstrated structural differences in intestinal glycosylation in TCRδ(-/-) mice compared with WT littermates. TCRδ(-/-) colons contained a lower proportion of core-2 structures and an increased proportion of core-1 structures whereas TCRδ(-/-) small intestines had a decreased percentage of core-3 structures. The glycan antennae in TCRδ(-/-) colon and small intestine showed altered structural diversity compared with WT mice. There were significant differences in the sialylated species between the TCRδ(-/-) and WT mice with the sialylated Tn antigen found exclusively in the TCRδ(-/-)small intestine, whereas the sulfation pattern remained mostly unchanged. These findings provide novel molecular insights underpinning the role of γδ IELs in maintaining gut homeostasis.


Asunto(s)
Mucosa Intestinal/metabolismo , Polisacáridos/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Ácidos Siálicos/metabolismo , Linfocitos T/metabolismo , Animales , Secuencia de Carbohidratos , Colon/citología , Colon/inmunología , Colon/metabolismo , Femenino , Expresión Génica , Glicosilación , Inmunidad Mucosa , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Especificidad de Órganos , Polisacáridos/química , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Ácidos Siálicos/química , Linfocitos T/citología , Linfocitos T/inmunología
4.
Cell Mol Gastroenterol Hepatol ; 14(2): 311-331, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35643188

RESUMEN

Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell-based in vitro models, often called organoids, and non-stem cell-based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases.


Asunto(s)
Organoides , Células Madre , Tracto Gastrointestinal , Estudios Prospectivos
5.
mSystems ; 7(4): e0149321, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35913188

RESUMEN

Serovars of the genus Salmonella primarily evolved as gastrointestinal pathogens in a wide range of hosts. Some serotypes later evolved further, adopting a more invasive lifestyle in a narrower host range associated with systemic infections. A system-level knowledge of these pathogens could identify the complex adaptations associated with the evolution of serovars with distinct pathogenicity, host range, and risk to human health. This promises to aid the design of interventions and serve as a knowledge base in the Salmonella research community. Here, we present SalmoNet2, a major update to SalmoNet1, the first multilayered interaction resource for Salmonella strains, containing protein-protein, transcriptional regulatory, and enzyme-enzyme interactions. The new version extends the number of Salmonella networks from 11 to 20. We now include a strain from the second species in the Salmonella genus, a strain from the Salmonella enterica subspecies arizonae and additional strains of importance from the subspecies enterica, including S. Typhimurium strain D23580, an epidemic multidrug-resistant strain associated with invasive nontyphoidal salmonellosis (iNTS). The database now uses strain specific metabolic models instead of a generalized model to highlight differences between strains. The update has increased the coverage of high-quality protein-protein interactions, and enhanced interoperability with other computational resources by adopting standardized formats. The resource website has been updated with tutorials to help researchers analyze their Salmonella data using molecular interaction networks from SalmoNet2. SalmoNet2 is accessible at http://salmonet.org/. IMPORTANCE Multilayered network databases collate interaction information from multiple sources, and are powerful both as a knowledge base and subject of analysis. Here, we present SalmoNet2, an integrated network resource containing protein-protein, transcriptional regulatory, and metabolic interactions for 20 Salmonella strains. Key improvements to the update include expanding the number of strains, strain-specific metabolic networks, an increase in high-quality protein-protein interactions, community standard computational formats to help interoperability, and online tutorials to help users analyze their data using SalmoNet2.


Asunto(s)
Infecciones por Salmonella , Salmonella enterica , Humanos , Salmonella/genética , Infecciones por Salmonella/epidemiología , Salmonella enterica/genética , Redes y Vías Metabólicas , Especificidad del Huésped
6.
Front Cell Infect Microbiol ; 12: 834895, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061866

RESUMEN

Macroautophagy is a ubiquitous homeostasis and health-promoting recycling process of eukaryotic cells, targeting misfolded proteins, damaged organelles and intracellular infectious agents. Some intracellular pathogens such as Salmonella enterica serovar Typhimurium hijack this process during pathogenesis. Here we investigate potential protein-protein interactions between host transcription factors and secreted effector proteins of Salmonella and their effect on host gene transcription. A systems-level analysis identified Salmonella effector proteins that had the potential to affect core autophagy gene regulation. The effect of a SPI-1 effector protein, SopE, that was predicted to interact with regulatory proteins of the autophagy process, was investigated to validate our approach. We then confirmed experimentally that SopE can directly bind to SP1, a host transcription factor, which modulates the expression of the autophagy gene MAP1LC3B. We also revealed that SopE might have a double role in the modulation of autophagy: Following initial increase of MAP1LC3B transcription triggered by Salmonella infection, subsequent decrease in MAP1LC3B transcription at 6h post-infection was SopE-dependent. SopE also played a role in modulation of the autophagy flux machinery, in particular MAP1LC3B and p62 autophagy proteins, depending on the level of autophagy already taking place. Upon typical infection of epithelial cells, the autophagic flux is increased. However, when autophagy was chemically induced prior to infection, SopE dampened the autophagic flux. The same was also observed when most of the intracellular Salmonella cells were not associated with the SCV (strain lacking sifA) regardless of the autophagy induction status before infection. We demonstrated how regulatory network analysis can be used to better characterise the impact of pathogenic effector proteins, in this case, Salmonella. This study complements previous work in which we had demonstrated that specific pathogen effectors can affect the autophagy process through direct interaction with autophagy proteins. Here we show that effector proteins can also influence the upstream regulation of the process. Such interdisciplinary studies can increase our understanding of the infection process and point out targets important in intestinal epithelial cell defense.


Asunto(s)
Infecciones por Salmonella , Salmonella typhimurium , Autofagia/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Humanos , Salmonella typhimurium/genética
7.
Front Immunol ; 12: 629193, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33732251

RESUMEN

Hyper-induction of pro-inflammatory cytokines, also known as a cytokine storm or cytokine release syndrome (CRS), is one of the key aspects of the currently ongoing SARS-CoV-2 pandemic. This process occurs when a large number of innate and adaptive immune cells activate and start producing pro-inflammatory cytokines, establishing an exacerbated feedback loop of inflammation. It is one of the factors contributing to the mortality observed with coronavirus 2019 (COVID-19) for a subgroup of patients. CRS is not unique to the SARS-CoV-2 infection; it was prevalent in most of the major human coronavirus and influenza A subtype outbreaks of the past two decades (H5N1, SARS-CoV, MERS-CoV, and H7N9). With a comprehensive literature search, we collected changing the cytokine levels from patients upon infection with the viral pathogens mentioned above. We analyzed published patient data to highlight the conserved and unique cytokine responses caused by these viruses. Our curation indicates that the cytokine response induced by SARS-CoV-2 is different compared to other CRS-causing respiratory viruses, as SARS-CoV-2 does not always induce specific cytokines like other coronaviruses or influenza do, such as IL-2, IL-10, IL-4, or IL-5. Comparing the collated cytokine responses caused by the analyzed viruses highlights a SARS-CoV-2-specific dysregulation of the type-I interferon (IFN) response and its downstream cytokine signatures. The map of responses gathered in this study could help specialists identify interventions that alleviate CRS in different diseases and evaluate whether they could be used in the COVID-19 cases.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , SARS-CoV-2/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Índice de Severidad de la Enfermedad , COVID-19/sangre , COVID-19/patología , COVID-19/virología , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/virología , Citocinas/sangre , Humanos , Inflamación/inmunología , Gripe Humana/sangre , Gripe Humana/virología , Síndrome Respiratorio Agudo Grave/sangre , Síndrome Respiratorio Agudo Grave/virología
8.
Cells ; 10(9)2021 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-34571891

RESUMEN

Intercellular communication mediated by cytokines is critical to the development of immune responses, particularly in the context of infectious and inflammatory diseases. By releasing these small molecular weight peptides, the source cells can influence numerous intracellular processes in the target cells, including the secretion of other cytokines downstream. However, there are no readily available bioinformatic resources that can model cytokine-cytokine interactions. In this effort, we built a communication map between major tissues and blood cells that reveals how cytokine-mediated intercellular networks form during homeostatic conditions. We collated the most prevalent cytokines from the literature and assigned the proteins and their corresponding receptors to source tissue and blood cell types based on enriched consensus RNA-Seq data from the Human Protein Atlas database. To assign more confidence to the interactions, we integrated the literature information on cell-cytokine interactions from two systems of immunology databases, immuneXpresso and ImmunoGlobe. From the collated information, we defined two metanetworks: a cell-cell communication network connected by cytokines; and a cytokine-cytokine interaction network depicting the potential ways in which cytokines can affect the activity of each other. Using expression data from disease states, we then applied this resource to reveal perturbations in cytokine-mediated intercellular signalling in inflammatory and infectious diseases (ulcerative colitis and COVID-19, respectively). For ulcerative colitis, with CytokineLink, we demonstrated a significant rewiring of cytokine-mediated intercellular communication between non-inflamed and inflamed colonic tissues. For COVID-19, we were able to identify cell types and cytokine interactions following SARS-CoV-2 infection, highlighting important cytokine interactions that might contribute to severe illness in a subgroup of patients. Such findings have the potential to inform the development of novel, cytokine-targeted therapeutic strategies. CytokineLink is freely available for the scientific community through the NDEx platform and the project github repository.


Asunto(s)
COVID-19/inmunología , Citocinas/metabolismo , Inmunidad , Enfermedades Inflamatorias del Intestino/inmunología , Comunicación Celular , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Bases de Datos Genéticas , Humanos , Enfermedades Inflamatorias del Intestino/patología , Transducción de Señal
9.
Gastroenterology ; 137(2): 579-87, 587.e1-2, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19375423

RESUMEN

BACKGROUND & AIMS: Intestinal dendritic cells (DCs) sample bacteria, such as Salmonella, by extending cellular processes into the lumen to capture bacteria and shuttle them across the epithelium; however, direct evidence of bacteria-loaded DCs travelling back into the tissue is lacking. We hypothesized that sampling is paralleled by migration of DCs into the lumen prior to or following the internalization of Salmonella. METHODS: The small intestine and the colon of BALB/c and C57BL/6 mice were challenged with noninvasive Salmonella enterica serovar Typhimurium SL1344-DeltaSalmonella pathogenicity island (SPI) 1 or Escherichia coli DH5alpha by using isolated loops or oral administration by gavage. Transepithelial migration of DCs was documented by immunohistochemistry, microscopy, and flow cytometry. The role of flagellin was determined by using flagellin (DeltafliC DeltafljB)- and SPI1-SPI2 (DeltaSPI1 DeltassrA)-deficient Salmonella, flagellated E coli K12, and MyD88 mice. RESULTS: Salmonella DeltaSPI1 induced migration of CD11c(+)CX(3)CR1(+)MHCII(+)CD11b(-)CD8alpha(-) DCs into the small intestine, whereas flagellin- and SPI1-SPI2-deficient Salmonella, soluble flagellin, and E coli DH5alpha or flagellated K12, failed to do so. DC migration did not occur in the colon; it was not observed in MyD88 mice, and intraluminal DCs internalized Salmonella but did not cross the epithelium to return into tissues. Finally, DC migration was not linked to Salmonella-induced damage of the epithelium. CONCLUSIONS: DC-mediated sampling of Salmonella is accompanied by flagellin- and MyD88-dependent migration of Salmonella-capturing DCs into the intestinal lumen. We suggest that the rapid intraluminal migration of Salmonella-capturing DCs may play a role in the protection of the intestinal mucosa against bacterial infection.


Asunto(s)
Movimiento Celular/fisiología , Células Dendríticas/citología , Flagelina/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Salmonella enterica/inmunología , Animales , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Movimiento Celular/inmunología , Colon/citología , Colon/microbiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Modelos Animales de Enfermedad , Femenino , Flagelina/inmunología , Citometría de Flujo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestino Delgado/citología , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Probabilidad , Distribución Aleatoria , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/fisiopatología , Sensibilidad y Especificidad
10.
Front Microbiol ; 11: 57, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117106

RESUMEN

Gram-negative bacteria ubiquitously produce and release nano-size, non-replicative outer membrane vesicles (OMVs). In the gastrointestinal (GI-) tract, OMVs generated by members of the intestinal microbiota are believed to contribute to maintaining the intestinal microbial ecosystem and mediating bacteria-host interactions, including the delivery of bacterial effector molecules to host cells to modulate their physiology. Bacterial OMVs have also been found in the bloodstream although their origin and fate are unclear. Here we have investigated the interactions between OMVs produced by the major human gut commensal bacterium, Bacteroides thetaiotaomicron (Bt), with cells of the GI-tract. Using a combination of in vitro culture systems including intestinal epithelial organoids and in vivo imaging we show that intestinal epithelial cells principally acquire Bt OMVs via dynamin-dependent endocytosis followed by intracellular trafficking to LAMP-1 expressing endo-lysosomal vesicles and co-localization with the perinuclear membrane. We observed that Bt OMVs can also transmigrate through epithelial cells via a paracellular route with in vivo imaging demonstrating that within hours of oral administration Bt OMVs can be detected in systemic tissues and in particular, the liver. Our findings raise the intriguing possibility that OMVs may act as a long-distance microbiota-host communication system.

11.
Autophagy ; 15(9): 1620-1633, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30909843

RESUMEN

Due to the critical role played by autophagy in pathogen clearance, pathogens have developed diverse strategies to subvert it. Despite previous key findings of bacteria-autophagy interplay, asystems-level insight into selective targeting by the host and autophagy modulation by the pathogens is lacking. We predicted potential interactions between human autophagy proteins and effector proteins from 56 pathogenic bacterial species by identifying bacterial proteins predicted to have recognition motifs for selective autophagy receptors SQSTM1/p62, CALCOCO2/NDP52 and MAP1LC3/LC3. Using structure-based interaction prediction, we identified bacterial proteins capable to modify core autophagy components. Our analysis revealed that autophagy receptors in general potentially target mostly genus-specific proteins, and not those present in multiple genera. The complementarity between the predicted SQSTM1/p62 and CALCOCO2/NDP52 targets, which has been shown for Salmonella, Listeria and Shigella, could be observed across other pathogens. This complementarity potentially leaves the host more susceptible to chronic infections upon the mutation of autophagy receptors. Proteins derived from enterotoxigenic and non-toxigenic Bacillus outer membrane vesicles indicated that autophagy targets pathogenic proteins rather than non-pathogenic ones. We also observed apathogen-specific pattern as to which autophagy phase could be modulated by specific genera. We found intriguing examples of bacterial proteins that could modulate autophagy, and in turn being targeted by autophagy as ahost defense mechanism. We confirmed experimentally an interplay between a Salmonella protease, YhjJ and autophagy. Our comparative meta-analysis points out key commonalities and differences in how pathogens could affect autophagy and how autophagy potentially recognizes these pathogenic effectors. Abbreviations: ATG5: autophagy related 5; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; GST: glutathione S-transferase; LIR: MAP1LC3/LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3 alpha; OMV: outer membrane vesicles; SQSTM1/p62: sequestosome 1; SCV: Salmonella containing vesicle; TECPR1: tectonin beta-propeller repeat containing 1; YhjJ: hypothetical zinc-protease.


Asunto(s)
Autofagia/genética , Proteínas Bacterianas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Proteína Sequestosoma-1/metabolismo , Secuencias de Aminoácidos , Bacillus/metabolismo , Bacillus/patogenicidad , Análisis por Conglomerados , Biología Computacional , Evolución Molecular , Interacciones Huésped-Patógeno/genética , Humanos , Listeria/metabolismo , Listeria/patogenicidad , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , Unión Proteica/genética , Mapas de Interacción de Proteínas , Salmonella/metabolismo , Salmonella/patogenicidad , Proteína Sequestosoma-1/genética , Shigella/metabolismo , Shigella/patogenicidad , Factores de Virulencia/metabolismo
12.
Dis Model Mech ; 12(3)2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30814064

RESUMEN

Paneth cells are key epithelial cells that provide an antimicrobial barrier and maintain integrity of the small-intestinal stem cell niche. Paneth cell abnormalities are unfortunately detrimental to gut health and are often associated with digestive pathologies such as Crohn's disease or infections. Similar alterations are observed in individuals with impaired autophagy, a process that recycles cellular components. The direct effect of autophagy impairment on Paneth cells has not been analysed. To investigate this, we generated a mouse model lacking Atg16l1 specifically in intestinal epithelial cells, making these cells impaired in autophagy. Using three-dimensional intestinal organoids enriched for Paneth cells, we compared the proteomic profiles of wild-type and autophagy-impaired organoids. We used an integrated computational approach combining protein-protein interaction networks, autophagy-targeted proteins and functional information to identify the mechanistic link between autophagy impairment and disrupted pathways. Of the 284 altered proteins, 198 (70%) were more abundant in autophagy-impaired organoids, suggesting reduced protein degradation. Interestingly, these differentially abundant proteins comprised 116 proteins (41%) that are predicted targets of the selective autophagy proteins p62, LC3 and ATG16L1. Our integrative analysis revealed autophagy-mediated mechanisms that degrade key proteins in Paneth cell functions, such as exocytosis, apoptosis and DNA damage repair. Transcriptomic profiling of additional organoids confirmed that 90% of the observed changes upon autophagy alteration have effects at the protein level, not on gene expression. We performed further validation experiments showing differential lysozyme secretion, confirming our computationally inferred downregulation of exocytosis. Our observations could explain how protein-level alterations affect Paneth cell homeostatic functions upon autophagy impairment.This article has an associated First Person interview with the joint first authors of the paper.


Asunto(s)
Autofagia , Intestinos/fisiología , Organoides/citología , Organoides/metabolismo , Células de Paneth/metabolismo , Proteómica , Transcriptoma/genética , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/metabolismo , Células Epiteliales/metabolismo , Exocitosis , Femenino , Masculino , Ratones Endogámicos C57BL , Proteolisis , Reproducibilidad de los Resultados
13.
Curr Opin Microbiol ; 7(3): 277-82, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15196496

RESUMEN

The understanding of bacterial pathogenesis is dependent on techniques that elucidate the underlying genetic and biochemical mechanisms. To study the mechanism of bacterial survival and proliferation within host cells we need accurate tools that tell us what is occurring within the infecting organism. It has now become possible to determine the transcriptional status of in vivo-derived bacteria at the level of the whole genome. Such expression profiles serve as a monitor of the host cell environment as well as an indicator of the bacterial adaptation to its intracellular niche. Here, we review the methods used to produce microarray data for defining the bacterial intracellular transcriptome, and examine the pitfalls in extracting bacterial RNA from the infected host compartment.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Proteínas Bacterianas/metabolismo , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Proteínas Bacterianas/genética , Línea Celular , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , ARN Bacteriano/análisis , ARN Bacteriano/aislamiento & purificación , Transcripción Genética
14.
Methods Enzymol ; 358: 43-66, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12474378

RESUMEN

To date, the majority of studies of bacterial gene expression have been carried out on large communities, as techniques for analysis of expression in individual cells have not been available. Recent developments now allow us to use reporter genes to monitor gene expression in individual bacterial cells. Conventional reporters are not suitable for studies of living single cells. However, variants of GFP have proved to be ideal for the study of development, cell biology, and pathogenesis and are now the reporters of choice for microbial studies. In combination with techniques such as DFI and IVET and the use of flow cytometry and advanced fluorescence microscopy, the latest generation of GFP reporters allows the investigation of gene expression in individual bacterial cells within particular environments. These studies promise to bring a new level of understanding to the fields of bacterial pathogenesis and environmental microbiology.


Asunto(s)
Proteínas Luminiscentes/genética , Salmonella typhimurium/genética , Sustitución de Aminoácidos , Animales , Técnicas Bacteriológicas/métodos , Citometría de Flujo/métodos , Genes Reporteros , Marcadores Genéticos , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/análisis , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/análisis , Reproducibilidad de los Resultados , Salmonella typhimurium/crecimiento & desarrollo , Escifozoos , Espectrometría de Fluorescencia/métodos
15.
Cell Rep ; 6(4): 646-56, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24529702

RESUMEN

Dietary InsP6 can modulate eukaryotic cell proliferation and has complex nutritive consequences, but its metabolism in the mammalian gastrointestinal tract is poorly understood. Therefore, we performed phylogenetic analyses of the gastrointestinal microbiome in order to search for candidate InsP6 phosphatases. We determined that prominent gut bacteria express homologs of the mammalian InsP6 phosphatase (MINPP) and characterized the enzyme from Bacteroides thetaiotaomicron (BtMinpp). We show that BtMinpp has exceptionally high catalytic activity, which we rationalize on the basis of mutagenesis studies and by determining its crystal structure at 1.9 Å resolution. We demonstrate that BtMinpp is packaged inside outer membrane vesicles (OMVs) protecting the enzyme from degradation by gastrointestinal proteases. Moreover, we uncover an example of cross-kingdom cell-to-cell signaling, showing that the BtMinpp-OMVs interact with intestinal epithelial cells to promote intracellular Ca(2+) signaling. Our characterization of BtMinpp offers several directions for understanding how the microbiome serves human gastrointestinal physiology.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroides/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bacteroides/química , Bacteroides/genética , Señalización del Calcio , Dominio Catalítico , Células HT29 , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Datos de Secuencia Molecular , Mutación , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/genética , Filogenia , Proteolisis
16.
PLoS One ; 8(12): e84553, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358364

RESUMEN

Antimicrobial proteins influence intestinal microbial ecology and limit proliferation of pathogens, yet the regulation of their expression has only been partially elucidated. Here, we have identified a putative pathway involving epithelial cells and intestinal intraepithelial lymphocytes (iIELs) that leads to antimicrobial protein (AMP) production by Paneth cells. Mice lacking γδ iIELs (TCRδ(-/-)) express significantly reduced levels of the AMP angiogenin 4 (Ang4). These mice were also unable to up-regulate Ang4 production following oral challenge by Salmonella, leading to higher levels of mucosal invasion compared to their wild type counterparts during the first 2 hours post-challenge. The transfer of γδ iIELs from wild type (WT) mice to TCRδ(-/-) mice restored Ang4 production and Salmonella invasion levels were reduced to those obtained in WT mice. The ability to restore Ang4 production in TCRδ(-/-) mice was shown to be restricted to γδ iIELs expressing Vγ7-encoded TCRs. Using a novel intestinal crypt co-culture system we identified a putative pathway of Ang4 production initiated by exposure to Salmonella, intestinal commensals or microbial antigens that induced intestinal epithelial cells to produce cytokines including IL­23 in a TLR-mediated manner. Exposure of TCR-Vγ7(+) γδ iIELs to IL-23 promoted IL­22 production, which triggered Paneth cells to secrete Ang4. These findings identify a novel role for γδ iIELs in mucosal defence through sensing immediate epithelial cell cytokine responses and influencing AMP production. This in turn can contribute to the maintenance of intestinal microbial homeostasis and epithelial barrier function, and limit pathogen invasion.


Asunto(s)
Comunicación Celular , Enterocitos/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos/metabolismo , Células de Paneth/metabolismo , Ribonucleasa Pancreática/biosíntesis , Animales , Línea Celular , Interleucina-23/biosíntesis , Interleucinas/farmacología , Mucosa Intestinal/microbiología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Salmonella/inmunología , Estrés Fisiológico , Interleucina-22
17.
Microbiology (Reading) ; 155(Pt 9): 2919-2929, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542004

RESUMEN

Toll-like receptor 4 (TLR4) senses bacterial LPS and is required for the control of systemic Salmonella enterica serovar Typhimurium infection in mice. The mechanisms of TLR4 activation and its downstream signalling cascades are well described, yet the direct effects on the pathogen of signalling via this receptor remain unknown. To investigate this we used microarray-based transcriptome profiling of intracellular S. Typhimurium during infection of primary bone marrow-derived macrophages from wild-type and TLR4-deficient mice. We identified 17 S. Typhimurium genes that were upregulated in the presence of functional TLR4. Nine of these genes have putative functions in oxidative stress resistance. We therefore examined S. Typhimurium gene expression during infection of NADPH oxidase-deficient macrophages, which lack normal oxidative killing mechanisms. We identified significant overlap between the 'TLR4-responsive' and 'NADPH oxidase-responsive' genes. This is new evidence for a link between TLR4 signalling and NADPH oxidase activity. Interestingly, with the exception of a dps mutant, S. Typhimurium strains lacking individual TLR4- and/or oxidative stress-responsive genes were not attenuated during intravenous murine infections. Our study shows that TLR4 activity, either directly or indirectly, induces the expression of multiple stress resistance genes during the intracellular life of S. Typhimurium.


Asunto(s)
Salmonelosis Animal/metabolismo , Salmonella typhimurium/fisiología , Transducción de Señal , Estrés Fisiológico/genética , Receptor Toll-Like 4/metabolismo , Animales , Técnicas de Cultivo de Célula , Perfilación de la Expresión Génica , Genes Bacterianos , Interacciones Huésped-Patógeno , Ratones , Estrés Oxidativo/genética , ARN Bacteriano/análisis , ARN Bacteriano/biosíntesis , ARN Bacteriano/genética , Receptores Inmunológicos/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/patogenicidad , Virulencia
18.
Cell Microbiol ; 9(2): 413-24, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16953802

RESUMEN

By combining intravital multiphoton microscopy and bacterial genetics we have developed a technique enabling real-time imaging of bacterial proliferation and tissue responses in a live animal. Spatial and temporal control of the infection process was achieved by microinjecting GFP(+)-expressing uropathogenic Escherichia coli (UPEC) into tubules of exteriorized kidneys in live rats. GFP(+) was introduced in the clinical UPEC strain CFT073 as a single-copy chromosomal gene fusion. Within hours, bacterial colonization was accompanied by marked ischaemic effects, perivascular leakage, loss of tubular integrity and localized recruitment of immune cells. The pathophysiology was altered in response to an isogenic bacterial strain lacking the exotoxin haemolysin, revealing the subtle and temporal roles of bacterial virulence factors in vivo. Microdissection and RNA extraction of the injected nephron allowed molecular analysis of prokaryotic and eukaryotic gene expression. The techniques described here can be applied to study the integrated cell communication evoked by a variety of bacterial pathogens, assisting in the design of strategies to combat bacterial infections.


Asunto(s)
Infecciones Bacterianas/patología , Sistemas de Computación , Progresión de la Enfermedad , Escherichia coli/patogenicidad , Animales , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Proteínas Fluorescentes Verdes , Procesamiento de Imagen Asistido por Computador , Peritonitis/microbiología , Peritonitis/patología , Ratas , Infecciones Urinarias/microbiología , Infecciones Urinarias/patología
19.
Appl Environ Microbiol ; 69(12): 7480-91, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14660401

RESUMEN

We developed a reliable and flexible green fluorescent protein (GFP)-based system for measuring gene expression in individual bacterial cells. Until now, most systems have relied upon plasmid-borne gfp gene fusions, risking problems associated with plasmid instability. We show that a recently developed GFP variant, GFP+, is suitable for assessing bacterial gene expression. Various gfp+ transcriptional fusions were constructed and integrated as single copies into the chromosome of Salmonella enterica serovar Typhimurium. A comparison of the expression levels of proU-lacZ and proU-gfp+ fusions showed that GFP+ reported proU activity in individual Salmonella cells as accurately as beta-galactosidase reported activity for entire populations. The single-copy gfp+ fusions were ideal for monitoring up- and downregulation of Salmonella virulence genes. We discovered that in vitro induction of the SPI1gene prgH occurs only in a portion of the population and that the proportion varies with the growth phase. We determined the level of expression of the SPI2 gene ssaG in bacteria released from murine macrophages. Our results demonstrate for the first time that single-copy GFP+ fusions reliably report gene expression in simple and complex environments. This approach promises to allow accurate measurement of gene expression in individual bacteria during animal infection.


Asunto(s)
Dosificación de Gen , Regulación Bacteriana de la Expresión Génica , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Macrófagos/microbiología , Ratones , Salmonella typhimurium/citología , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA