Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 278
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 48(6): 1233-1244.e6, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29858013

RESUMEN

Shigella is a Gram-negative bacterium that causes bacillary dysentery worldwide. It invades the intestinal epithelium to elicit intense inflammation and tissue damage, yet the underlying mechanisms of its host selectivity and low infectious inoculum remain perplexing. Here, we report that Shigella co-opts human α-defensin 5 (HD5), a host defense peptide important for intestinal homeostasis and innate immunity, to enhance its adhesion to and invasion of mucosal tissues. HD5 promoted Shigella infection in vitro in a structure-dependent manner. Shigella, commonly devoid of an effective host-adhesion apparatus, preferentially targeted HD5 to augment its ability to colonize the intestinal epithelium through interactions with multiple bacterial membrane proteins. HD5 exacerbated infectivity and Shigella-induced pathology in a culture of human colorectal tissues and three animal models. Our findings illuminate how Shigella exploits innate immunity by turning HD5 into a virulence factor for infection, unveiling a mechanism of action for this highly proficient human pathogen.


Asunto(s)
Adhesión Bacteriana/fisiología , Disentería Bacilar/inmunología , Interacciones Huésped-Patógeno/fisiología , Shigella/patogenicidad , alfa-Defensinas , Animales , Humanos
2.
Proc Natl Acad Sci U S A ; 119(41): e2209589119, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36197997

RESUMEN

Environmental enteric dysfunction (EED) is an inflammatory syndrome postulated to contribute to stunted child growth and to be associated with intestinal dysbiosis and nutrient malabsorption. However, the small intestinal contributions to EED remain poorly understood. This study aimed to assess changes in the proximal and distal intestinal microbiota in the context of stunting and EED and to test for a causal role of these bacterial isolates in the underlying pathophysiology. We performed a cross-sectional study in two African countries recruiting roughly 1,000 children aged 2 to 5 years and assessed the microbiota in the stomach, duodenum, and feces. Upper gastrointestinal samples were obtained from stunted children and stratified according to stunting severity. Fecal samples were collected. We then investigated the role of clinical isolates in EED pathophysiology using tissue culture and animal models. We find that small intestinal bacterial overgrowth (SIBO) is extremely common (>80%) in stunted children. SIBO is frequently characterized by an overgrowth of oral bacteria, leading to increased permeability and inflammation and to replacement of classical small intestinal strains. These duodenal bacterial isolates decrease lipid absorption in both cultured enterocytes and mice, providing a mechanism by which they may exacerbate EED and stunting. Further, we find a specific fecal signature associated with the EED markers fecal calprotectin and alpha-antitrypsin. Our study shows a causal implication of ectopic colonization of oral bacterial isolated from the small intestine in nutrient malabsorption and gut leakiness in vitro. These findings have important therapeutic implications for modulating the microbiota through microbiota-targeted interventions.


Asunto(s)
Microbioma Gastrointestinal , Trastornos del Crecimiento , Intestino Delgado , Lípidos , Boca , Animales , Bacterias , Preescolar , Estudios Transversales , Trastornos del Crecimiento/etiología , Humanos , Complejo de Antígeno L1 de Leucocito , Metabolismo de los Lípidos , Síndromes de Malabsorción , Ratones , Modelos Teóricos , Boca/microbiología
3.
EMBO J ; 39(23): e107227, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33205839

RESUMEN

A number of promising COVID-19 vaccine candidates may pass approval this month. However, the pandemic will only be brought into check through an equitable, epidemiologically informed distribution policy. The health emergency provides a unique opportunity for a new paradigm to mitigate between global health, national and commercial interests.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19/prevención & control , Industria Farmacéutica/economía , Programas de Inmunización/organización & administración , Vacunas contra la COVID-19/economía , Bases de Datos Factuales , Países en Desarrollo , Europa (Continente) , Salud Global/economía , Humanos , Programas de Inmunización/economía , Japón , Estados Unidos , Organización Mundial de la Salud
4.
Cell ; 138(3): 416-20, 2009 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-19665961

RESUMEN

Research on microbe-host interactions focuses principally on pathogens, yet our immune system must deal with the huge number of beneficial commensal bacteria in our gut. It is becoming clear that the host immune system must reach a delicate balance between destroying dangerous bacterial pathogens while preserving the beneficial gut microbiota.


Asunto(s)
Bacterias/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Animales , Humanos , Tolerancia Inmunológica , Probióticos
5.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34504012

RESUMEN

The interleukin-2 receptor (IL-2R) is a cytokine receptor essential for immunity that transduces proliferative signals regulated by its uptake and degradation. IL-2R is a well-known marker of clathrin-independent endocytosis (CIE), a process devoid of any coat protein, raising the question of how the CIE vesicle is generated. Here, we investigated the impact of IL-2Rγ clustering in its endocytosis. Combining total internal reflection fluorescence (TIRF) live imaging of a CRISPR-edited T cell line endogenously expressing IL-2Rγ tagged with green fluorescent protein (GFP), with multichannel imaging, single-molecule tracking, and quantitative analysis, we were able to decipher IL-2Rγ stoichiometry at the plasma membrane in real time. We identified three distinct IL-2Rγ cluster populations. IL-2Rγ is secreted to the cell surface as a preassembled small cluster of three molecules maximum, rapidly diffusing at the plasma membrane. A medium-sized cluster composed of four to six molecules is key for IL-2R internalization and is promoted by interleukin 2 (IL-2) binding, while larger clusters (more than six molecules) are static and inefficiently internalized. Moreover, we identified membrane cholesterol and the branched actin cytoskeleton as key regulators of IL-2Rγ clustering and IL-2-induced signaling. Both cholesterol depletion and Arp2/3 inhibition lead to the assembly of large IL-2Rγ clusters, arising from the stochastic interaction of receptor molecules in close correlation with their enhanced lateral diffusion at the membrane, thus resulting in a default in IL-2R endocytosis. Despite similar clustering outcomes, while cholesterol depletion leads to a sustained IL-2-dependent signaling, Arp2/3 inhibition prevents signal initiation. Taken together, our results reveal the importance of cytokine receptor clustering for CIE initiation and signal transduction.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Endocitosis , Receptores de Interleucina-2/metabolismo , Linfocitos T/metabolismo , Transporte Biológico , Humanos , Transducción de Señal
6.
Nat Immunol ; 12(2): 121-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21245903

RESUMEN

The last 10 years have witnessed the identification of a new class of intracellular pattern-recognition molecules--the nucleotide-binding domain and leucine-rich repeat-containing family (NLR). Members of this family garnered interest as pattern-recognition receptors able to trigger inflammatory responses against pathogens. Many studies support a pathogen-recognition function for human NLR proteins and shed light on their role in the broader control of adaptive immunity and various disease states. Other evidence suggests that NLRs function in processes unrelated to pathogen detection. Here we discuss recent advances in our understanding of the biology of the human NLR proteins and their non-pathogen-recognition function in tissue homeostasis, apoptosis, graft-versus-host disease and early development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Portadoras/inmunología , Enfermedad Injerto contra Huésped/inmunología , Células TH1/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis , Enfermedades Autoinmunes/genética , Proteínas Portadoras/genética , Desarrollo Embrionario/inmunología , Predisposición Genética a la Enfermedad , Humanos , Activación de Linfocitos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Ovulación/inmunología , Polimorfismo Genético , Transducción de Señal/inmunología , Activación Transcripcional/inmunología , Inmunología del Trasplante
7.
Proc Natl Acad Sci U S A ; 117(4): 1994-2003, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31919280

RESUMEN

The nucleotide-binding oligomerization domain-containing protein 2 (NOD2) agonist muramyl dipeptide (MDP), a peptidoglycan motif common to all bacteria, supports leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5)+ intestinal stem cell (ISC) survival through NOD2 activation upon an otherwise lethal oxidative stress-mediated signal. However, the underlying protective mechanisms remain unknown. Here, using irradiation as stressor and primarily murine-derived intestinal organoids as a model system, we show that MDP induced a significant reduction of total and mitochondrial reactive oxygen species (ROS) within ISCs, which was associated with mitophagy induction. ATG16L1 knockout (KO) and NOD2 KO organoids did not benefit from the MDP-induced cytoprotection. We confirmed the MDP-dependent induction of ISC mitophagy upon stress in vivo. These findings elucidate the NOD2-mediated mechanism of cytoprotection involving the clearance of the lethal excess of ROS molecules through mitophagy, triggered by the coordinated activation of NOD2 and ATG16L1 by a nuclear factor κB (NF-κB)-independent pathway.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/farmacología , Citoprotección , Intestinos/crecimiento & desarrollo , Mitofagia/efectos de los fármacos , Proteína Adaptadora de Señalización NOD2/metabolismo , Especies Reactivas de Oxígeno , Células Madre/citología , Animales , Células Cultivadas , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Inmunidad Innata/efectos de la radiación , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Mitofagia/efectos de la radiación , Proteína Adaptadora de Señalización NOD2/genética , Estrés Oxidativo/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/efectos de la radiación
8.
Gut ; 71(2): 296-308, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33593807

RESUMEN

OBJECTIVE: Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic ß-cells producing insulin. Both T1D patients and animal models exhibit gut microbiota and mucosa alterations, although the exact cause for these remains poorly understood. We investigated the production of key cytokines controlling gut integrity, the abundance of segmented filamentous bacteria (SFB) involved in the production of these cytokines, and the respective role of autoimmune inflammation and hyperglycaemia. DESIGN: We used several mouse models of autoimmune T1D as well as mice rendered hyperglycaemic without inflammation to study gut mucosa and microbiota dysbiosis. We analysed cytokine expression in immune cells, epithelial cell function, SFB abundance and microbiota composition by 16S sequencing. We assessed the role of anti-tumour necrosis factor α on gut mucosa inflammation and T1D onset. RESULTS: We show in models of autoimmune T1D a conserved loss of interleukin (IL)-17A, IL-22 and IL-23A in gut mucosa. Intestinal epithelial cell function was altered and gut integrity was impaired. These defects were associated with dysbiosis including progressive loss of SFB. Transfer of diabetogenic T-cells recapitulated these gut alterations, whereas induction of hyperglycaemia with no inflammation failed to do so. Moreover, anti-inflammatory treatment restored gut mucosa and immune cell function and dampened diabetes incidence. CONCLUSION: Our results demonstrate that gut mucosa alterations and dysbiosis in T1D are primarily linked to inflammation rather than hyperglycaemia. Anti-inflammatory treatment preserves gut homeostasis and protective commensal flora reducing T1D incidence.


Asunto(s)
Bacterias/aislamiento & purificación , Diabetes Mellitus Tipo 1/complicaciones , Disbiosis/etiología , Microbioma Gastrointestinal , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Animales , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Hiperglucemia/etiología , Inflamación/etiología , Mucosa Intestinal/metabolismo , Ratones
9.
EMBO J ; 37(23)2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30389666

RESUMEN

While mucosal inflammation is a major source of stress during enteropathogen infection, it remains to be fully elucidated how the host benefits from this environment to clear the pathogen. Here, we show that host stress induced by different stimuli mimicking inflammatory conditions strongly reduces the binding of Shigella flexneri to epithelial cells. Mechanistically, stress activates acid sphingomyelinase leading to host membrane remodeling. Consequently, knockdown or pharmacological inhibition of the acid sphingomyelinase blunts the stress-dependent inhibition of Shigella binding to host cells. Interestingly, stress caused by intracellular Shigella replication also results in remodeling of the host cell membrane, in vitro and in vivo, which precludes re-infection by this and other non-motile pathogens. In contrast, Salmonella Typhimurium overcomes the shortage of permissive entry sites by gathering effectively at the remaining platforms through its flagellar motility. Overall, our findings reveal host membrane remodeling as a novel stress-responsive cell-autonomous defense mechanism that protects epithelial cells from infection by non-motile bacterial pathogens.


Asunto(s)
Membrana Celular/inmunología , Disentería Bacilar/inmunología , Células Epiteliales/inmunología , Inmunidad Innata , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Shigella flexneri/inmunología , Estrés Fisiológico/inmunología , Animales , Membrana Celular/patología , Disentería Bacilar/patología , Células Epiteliales/patología , Cobayas , Infecciones por Salmonella/patología
10.
Immunity ; 39(6): 1121-31, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24332032

RESUMEN

Upon infection with Shigella flexneri, epithelial cells release ATP through connexin hemichannels. However, the pathophysiological consequence and the regulation of this process are unclear. Here we showed that in intestinal epithelial cell ATP release was an early alert response to infection with enteric pathogens that eventually promoted inflammation of the gut. Shigella evolved to escape this inflammatory reaction by its type III secretion effector IpgD, which blocked hemichannels via the production of the lipid PtdIns5P. Infection with an ipgD mutant resulted in rapid hemichannel-dependent accumulation of extracellular ATP in vitro and in vivo, which preceded the onset of inflammation. At later stages of infection, ipgD-deficient Shigella caused strong intestinal inflammation owing to extracellular ATP. We therefore describe a new paradigm of host-pathogen interaction based on endogenous danger signaling and identify extracellular ATP as key regulator of mucosal inflammation during infection. Our data provide new angles of attack for the development of anti-inflammatory molecules.


Asunto(s)
Adenosina Trifosfato/metabolismo , Disentería Bacilar/inmunología , Disentería Bacilar/metabolismo , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Shigella flexneri/metabolismo , Animales , Células Cultivadas , Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Células HeLa , Humanos , Mucosa Intestinal/patología , Masculino , Fosfatos de Fosfatidilinositol/genética , Reacción en Cadena de la Polimerasa , Conejos , Shigella flexneri/genética
11.
BMC Public Health ; 22(1): 1320, 2022 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-35810292

RESUMEN

BACKGROUND: Anaemia occurs in children when the haemoglobin level in the blood is less than the normal (11 g/dL), the consequence is the decrease of oxygen quantity in the tissues. It is a prevalent public health problem in many low-income countries, including Madagascar, and data on risk factors are lacking. We used existing data collected within the pathophysiology of environmental enteric dysfunction (EED) in Madagascar and the Central African Republic project (AFRIBIOTA project) conducted in underprivileged neighbourhoods of Antananarivo to investigate the factors associated with anaemia in children 24 to 59 months of age. METHODS: Children included in the AFRIBIOTA project in Antananarivo for whom data on haemoglobin and ferritin concentrations were available were included in the study. Logistic regression modelling was performed to identify factors associated with anaemia. RESULTS: Of the 414 children included in this data analysis, 24.4% were found to suffer from anaemia. We found that older children (adjusted OR: 0.95; 95% CI: 0.93-0.98) were less likely to have anaemia. Those with iron deficiency (adjusted OR: 6.1; 95% CI: 3.4-11.1) and those with a high level of faecal calprotectin (adjusted OR: 2.5; 95% CI: 1.4-4.4) were more likely to have anaemia than controls. CONCLUSIONS: To reduce anaemia in the children in this underprivileged area, more emphasis should be given to national strategies that improve children's dietary quality and micronutrient intake. Furthermore, existing measures should be broadened to include measures to reduce infectious disease burden.


Asunto(s)
Anemia Ferropénica , Anemia , Deficiencias de Hierro , Adolescente , Anemia/epidemiología , Anemia Ferropénica/epidemiología , Niño , Preescolar , Ferritinas , Humanos , Madagascar/epidemiología , Pobreza , Prevalencia
12.
Proc Natl Acad Sci U S A ; 116(48): 24285-24295, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31712445

RESUMEN

Sporadic colorectal cancer (CRC) is a result of complex interactions between the host and its environment. Environmental stressors act by causing host cell DNA alterations implicated in the onset of cancer. Here we investigate the stressor ability of CRC-associated gut dysbiosis as causal agent of host DNA alterations. The epigenetic nature of these alterations was investigated in humans and in mice. Germ-free mice receiving fecal samples from subjects with normal colonoscopy or from CRC patients were monitored for 7 or 14 wk. Aberrant crypt foci, luminal microbiota, and DNA alterations (colonic exome sequencing and methylation patterns) were monitored following human feces transfer. CRC-associated microbiota induced higher numbers of hypermethylated genes in murine colonic mucosa (vs. healthy controls' microbiota recipients). Several gene promoters including SFRP1,2,3, PENK, NPY, ALX4, SEPT9, and WIF1 promoters were found hypermethylated in CRC but not in normal tissues or effluents from fecal donors. In a pilot study (n = 266), the blood methylation levels of 3 genes (Wif1, PENK, and NPY) were shown closely associated with CRC dysbiosis. In a validation study (n = 1,000), the cumulative methylation index (CMI) of these genes was significantly higher in CRCs than in controls. Further, CMI appeared as an independent risk factor for CRC diagnosis as shown by multivariate analysis that included fecal immunochemical blood test. Consequently, fecal bacterial species in individuals with higher CMI in blood were identified by whole metagenomic analysis. Thus, CRC-related dysbiosis induces methylation of host genes, and corresponding CMIs together with associated bacteria are potential biomarkers for CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Epigénesis Genética , Microbioma Gastrointestinal/genética , Animales , Estudios de Cohortes , Metilación de ADN , Disbiosis/genética , Disbiosis/microbiología , Disbiosis/patología , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Regulación de la Expresión Génica , Vida Libre de Gérmenes , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones Endogámicos C3H , Regiones Promotoras Genéticas , ARN Ribosómico 16S
13.
Proc Natl Acad Sci U S A ; 116(27): 13582-13591, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31209035

RESUMEN

Intracellular trafficking pathways in eukaryotic cells are essential to maintain organelle identity and structure, and to regulate cell communication with its environment. Shigella flexneri invades and subverts the human colonic epithelium by the injection of virulence factors through a type 3 secretion system (T3SS). In this work, we report the multiple effects of two S. flexneri effectors, IpaJ and VirA, which target small GTPases of the Arf and Rab families, consequently inhibiting several intracellular trafficking pathways. IpaJ and VirA induce large-scale impairment of host protein secretion and block the recycling of surface receptors. Moreover, these two effectors decrease clathrin-dependent and -independent endocytosis. Therefore, S. flexneri infection induces a global blockage of host cell intracellular transport, affecting the exchange between cells and their external environment. The combined action of these effectors disorganizes the epithelial cell polarity, disturbs epithelial barrier integrity, promotes multiple invasion events, and enhances the pathogen capacity to penetrate into the colonic tissue in vivo.


Asunto(s)
Disentería Bacilar/fisiopatología , Mucosa Intestinal/microbiología , Shigella flexneri , Transporte Biológico , Células CACO-2 , Polaridad Celular , Colon/metabolismo , Colon/microbiología , Colon/patología , Colon/fisiopatología , Disentería Bacilar/metabolismo , Disentería Bacilar/patología , Endocitosis , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología
14.
EMBO J ; 36(17): 2567-2580, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701483

RESUMEN

The role of second messengers in the diversion of cellular processes by pathogens remains poorly studied despite their importance. Among these, Ca2+ virtually regulates all known cell processes, including cytoskeletal reorganization, inflammation, or cell death pathways. Under physiological conditions, cytosolic Ca2+ increases are transient and oscillatory, defining the so-called Ca2+ code that links cell responses to specific Ca2+ oscillatory patterns. During cell invasion, Shigella induces atypical local and global Ca2+ signals. Here, we show that by hydrolyzing phosphatidylinositol-(4,5)bisphosphate, the Shigella type III effector IpgD dampens inositol-(1,4,5)trisphosphate (InsP3) levels. By modifying InsP3 dynamics and diffusion, IpgD favors the elicitation of long-lasting local Ca2+ signals at Shigella invasion sites and converts Shigella-induced global oscillatory responses into erratic responses with atypical dynamics and amplitude. Furthermore, IpgD eventually inhibits InsP3-dependent responses during prolonged infection kinetics. IpgD thus acts as a pathogen regulator of the Ca2+ code implicated in a versatility of cell functions. Consistent with this function, IpgD prevents the Ca2+-dependent activation of calpain, thereby preserving the integrity of cell adhesion structures during the early stages of infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Calcio/metabolismo , Disentería Bacilar/metabolismo , Interacciones Huésped-Patógeno , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Shigella flexneri/fisiología , Calpaína/metabolismo , Adhesión Celular , Células HeLa , Humanos , Transducción de Señal
15.
Cell Microbiol ; 22(5): e13166, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31957253

RESUMEN

Strategies employed by pathogenic enteric bacteria, such as Shigella, to subvert the host adaptive immunity are not well defined. Impairment of T lymphocyte chemotaxis by blockage of polarised edge formation has been reported upon Shigella infection. However, the functional impact of Shigella on T lymphocytes remains to be determined. Here, we show that Shigella modulates CD4+ T cell F-actin dynamics and increases cell cortical stiffness. The scanning ability of T lymphocytes when encountering antigen-presenting cells (APC) is subsequently impaired resulting in decreased cell-cell contacts (or conjugates) between the two cell types, as compared with non-infected T cells. In addition, the few conjugates established between the invaded T cells and APCs display no polarised delivery and accumulation of the T cell receptor to the contact zone characterising canonical immunological synapses. This is most likely due to the targeting of intracellular vesicular trafficking by the bacterial type III secretion system (T3SS) effectors IpaJ and VirA. The collective impact of these cellular reshapings by Shigella eventually results in T cell activation dampening. Altogether, these results highlight the combined action of T3SS effectors leading to T cell defects upon Shigella infection.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Inmunidad Adaptativa , Disentería Bacilar/inmunología , Transporte de Proteínas/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Shigella/metabolismo , Actinas , Línea Celular , Aparato de Golgi , Humanos , Sinapsis Inmunológicas , Shigella/genética , Linfocitos T/inmunología , Sistemas de Secreción Tipo III/metabolismo
16.
Nature ; 520(7545): 99-103, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25600271

RESUMEN

The gut microbiota plays a crucial role in the maturation of the intestinal mucosal immune system of its host. Within the thousand bacterial species present in the intestine, the symbiont segmented filamentous bacterium (SFB) is unique in its ability to potently stimulate the post-natal maturation of the B- and T-cell compartments and induce a striking increase in the small-intestinal Th17 responses. Unlike other commensals, SFB intimately attaches to absorptive epithelial cells in the ileum and cells overlying Peyer's patches. This colonization does not result in pathology; rather, it protects the host from pathogens. Yet, little is known about the SFB-host interaction that underlies the important immunostimulatory properties of SFB, because SFB have resisted in vitro culturing for more than 50 years. Here we grow mouse SFB outside their host in an SFB-host cell co-culturing system. Single-celled SFB isolated from monocolonized mice undergo filamentation, segmentation, and differentiation to release viable infectious particles, the intracellular offspring, which can colonize mice to induce signature immune responses. In vitro, intracellular offspring can attach to mouse and human host cells and recruit actin. In addition, SFB can potently stimulate the upregulation of host innate defence genes, inflammatory cytokines, and chemokines. In vitro culturing thereby mimics the in vivo niche, provides new insights into SFB growth requirements and their immunostimulatory potential, and makes possible the investigation of the complex developmental stages of SFB and the detailed dissection of the unique SFB-host interaction at the cellular and molecular levels.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Técnicas de Cocultivo/métodos , Intestinos/inmunología , Intestinos/microbiología , Linfocitos/inmunología , Simbiosis/inmunología , Actinas/metabolismo , Animales , Bacterias/citología , Línea Celular , Escherichia coli/citología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Heces/microbiología , Femenino , Vida Libre de Gérmenes , Humanos , Inmunidad Mucosa/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/citología , Linfocitos/citología , Masculino , Ratones , Viabilidad Microbiana , Ganglios Linfáticos Agregados/inmunología , Células Th17/inmunología
17.
Matern Child Health J ; 25(10): 1626-1637, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34383227

RESUMEN

OBJECTIVES: With a fourth of all under-five children affected, stunting remains one of the biggest health challenges worldwide. Even though the main underlying factors are known, the exact pathways to stunting varying in affected regions, and interventions thus need to be tailored to the local contexts. This study aimed assessing and comparing factors associated with stunting in two understudied sub-Saharan urban contexts with some of the highest stunting prevalence globally: Bangui, Central African Republic (~ 36%) and Antananarivo, Madagascar (42%). METHODS: We performed a case-control study on 175 + 194 stunted and 237 + 230 non-stunted control children aged 2-5 years and matched for age, gender and district of residency. Factors associated with stunting were identified using a standardized, paper questionnaire delivered by trained interviewers. Statistical analysis was done using logistic regression modelling. RESULTS: In both sites, formal maternal education lowered the risk of being stunted and restricted access to soap, suffering of anaemia and low birth weight were associated with higher risk of stunting. Short maternal stature, household head different from parents, diarrhoea and coughing were associated with an increased risk and continuing breastfeeding was associated with a lower risk of stunting in Antananarivo. Previous severe undernutrition and dermatitis/ fungal skin infections were associated with higher and changes in diet during pregnancy with lower risk of stunting in Bangui. CONCLUSIONS: Our results suggest maternal education, antenatal care, iron supplementation and simple WASH interventions such as using soap and infection control as general and breastfeeding (Antananarivo) or better nutrition (Bangui) as area-specified interventions.


Asunto(s)
Trastornos del Crecimiento , Estado Nutricional , Estudios de Casos y Controles , República Centroafricana/epidemiología , Niño , Preescolar , Femenino , Trastornos del Crecimiento/epidemiología , Trastornos del Crecimiento/etiología , Humanos , Lactante , Madagascar/epidemiología , Embarazo , Prevalencia , Factores de Riesgo
18.
Proc Natl Acad Sci U S A ; 115(42): E9869-E9878, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30275324

RESUMEN

Antimicrobial peptides (AMPs) are mucosal defense effectors of the human innate immune response. In the intestine, AMPs are produced and secreted by epithelial cells to protect the host against pathogens and to support homeostasis with commensals. The inducible nature of AMPs suggests that potent inducers could be used to increase their endogenous expression for the prevention or treatment of diseases. Here we aimed at identifying molecules from the natural pharmacopoeia that induce expression of human ß-defensin-3 (HBD3), one of the most efficient AMPs, without modifying the production of proinflammatory cytokines. By screening, we identified three molecules isolated from medicinal plants, andrographolide, oridonin, and isoliquiritigenin, which induced HBD3 production in human colonic epithelial cells. This effect was observed without activation of the NF-κB pathway or the expression of associated proinflammatory cytokines. We identified the EGF receptor as the target of these compounds and characterized the downstream-activated MAPK pathways. At the chromatin level, molecules increased phosphorylation of histone H3 on serine S10 and recruitment of the c-Fos, c-Jun, and Elk1 or c-Myc transcription factors at the HBD3 promoter. Interestingly, stimulating cells with a combination of andrographolide and isoliquiritigenin synergistically enhanced HBD3 induction 10-fold more than observed with each molecule alone. Finally, we investigated the molecular basis governing the synergistic effect, confirmed our findings in human colonic primary cells, and demonstrated that synergism increased cellular antimicrobial activity. This work shows the capability of small molecules to achieve induction of epithelial antimicrobial defenses while simultaneously avoiding the deleterious risks of an inflammatory response.


Asunto(s)
Antiinfecciosos/farmacología , Bacterias/efectos de los fármacos , Productos Biológicos/farmacología , Colon/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , beta-Defensinas/metabolismo , Antiinflamatorios/farmacología , Células Cultivadas , Chalconas/farmacología , Colon/citología , Colon/efectos de los fármacos , Diterpenos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos
19.
Proc Natl Acad Sci U S A ; 115(2): E283-E291, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29279402

RESUMEN

Colonization by Streptococcus gallolyticus subsp. gallolyticus (SGG) is strongly associated with the occurrence of colorectal cancer (CRC). However, the factors leading to its successful colonization are unknown, and whether SGG influences the oncogenic process or benefits from the tumor-prone environment to prevail remains an open question. Here, we elucidate crucial steps that explain how CRC favors SGG colonization. By using mice genetically prone to CRC, we show that SGG colonization is 1,000-fold higher in tumor-bearing mice than in normal mice. This selective advantage occurs at the expense of resident intestinal enterococci. An SGG-specific locus encoding a bacteriocin ("gallocin") is shown to kill enterococci in vitro. Importantly, bile acids strongly enhance this bacteriocin activity in vivo, leading to greater SGG colonization. Constitutive activation of the Wnt pathway, one of the earliest signaling alterations in CRC, and the decreased expression of the bile acid apical transporter gene Slc10A2, as an effect of the Apc founding mutation, may thereby sustain intestinal colonization by SGG. We conclude that CRC-specific conditions promote SGG colonization of the gut by replacing commensal enterococci in their niche.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Tracto Gastrointestinal/microbiología , Streptococcus gallolyticus/fisiología , Adenoma , Animales , Bacteriocinas/genética , Bacteriocinas/metabolismo , Ácidos y Sales Biliares/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Simportadores/genética , Simportadores/metabolismo
20.
Proc Natl Acad Sci U S A ; 115(36): E8489-E8498, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30126990

RESUMEN

Linear growth delay (stunting) affects roughly 155 million children under the age of 5 years worldwide. Treatment has been limited by a lack of understanding of the underlying pathophysiological mechanisms. Stunting is most likely associated with changes in the microbial community of the small intestine, a compartment vital for digestion and nutrient absorption. Efforts to better understand the pathophysiology have been hampered by difficulty of access to small intestinal fluids. Here, we describe the microbial community found in the upper gastrointestinal tract of stunted children aged 2-5 y living in sub-Saharan Africa. We studied 46 duodenal and 57 gastric samples from stunted children, as well as 404 fecal samples from stunted and nonstunted children living in Bangui, Central African Republic, and in Antananarivo, Madagascar, using 16S Illumina Amplicon sequencing and semiquantitative culture methods. The vast majority of the stunted children showed small intestinal bacterial overgrowth dominated by bacteria that normally reside in the oropharyngeal cavity. There was an overrepresentation of oral bacteria in fecal samples of stunted children, opening the way for developing noninvasive diagnostic markers. In addition, Escherichia coli/Shigella sp. and Campylobacter sp. were found to be more prevalent in stunted children, while Clostridia, well-known butyrate producers, were reduced. Our data suggest that stunting is associated with a microbiome "decompartmentalization" of the gastrointestinal tract characterized by an increased presence of oropharyngeal bacteria from the stomach to the colon, hence challenging the current view of stunting arising solely as a consequence of small intestine overstimulation through recurrent infections by enteric pathogens.


Asunto(s)
Campylobacter , Desarrollo Infantil , Clostridium , Escherichia coli , Microbioma Gastrointestinal , Trastornos del Crecimiento , Intestino Delgado , Shigella , Campylobacter/clasificación , Campylobacter/aislamiento & purificación , Campylobacter/metabolismo , Preescolar , Clostridium/clasificación , Clostridium/aislamiento & purificación , Clostridium/metabolismo , Escherichia coli/clasificación , Escherichia coli/aislamiento & purificación , Escherichia coli/metabolismo , Femenino , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/microbiología , Humanos , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Masculino , Shigella/clasificación , Shigella/aislamiento & purificación , Shigella/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA