Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(3): 321-333, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135254

RESUMEN

The FcµR receptor for the crystallizable fragment (Fc) of immunoglobulin M (IgM) can function as a cell-surface receptor for secreted IgM on a variety of cell types. We found here that FcµR was also expressed in the trans-Golgi network of developing B cells, where it constrained transport of the IgM-isotype BCR (IgM-BCR) but not of the IgD-isotype BCR (IgD-BCR). In the absence of FcµR, the surface expression of IgM-BCR was increased, which resulted in enhanced tonic BCR signaling. B-cell-specific deficiency in FcµR enhanced the spontaneous differentiation of B-1 cells, which resulted in increased serum concentrations of natural IgM and dysregulated homeostasis of B-2 cells; this caused the spontaneous formation of germinal centers, increased titers of serum autoantibodies and excessive accumulation of B cells. Thus, FcµR serves as a critical regulator of B cell biology by constraining the transport and cell-surface expression of IgM-BCR.


Asunto(s)
Linfocitos B/fisiología , Inmunoglobulina M/metabolismo , Células Precursoras de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores Fc/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica , Inmunoglobulina M/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal , Células TH1/inmunología , Células Th2/inmunología
2.
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
3.
Proc Natl Acad Sci U S A ; 120(47): e2312453120, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37956278

RESUMEN

To mediate critical host-microbe interactions in the human small intestine, Paneth cells constitutively produce abundant levels of α-defensins and other antimicrobials. We report that the expression profile of these antimicrobials is dramatically askew in human small intestinal organoids (enteroids) as compared to that in paired tissue from which they are derived, with a reduction of α-defensins to nearly undetectable levels. Murine enteroids, however, recapitulate the expression profile of Paneth cell α-defensins seen in tissue. WNT/TCF signaling has been found to be instrumental in the regulation of α-defensins, yet in human enteroids exogenous stimulation of WNT signaling appears insufficient to rescue α-defensin expression. By stark contrast, forkhead box O (FOXO) inhibitor AS1842856 induced the expression of α-defensin mRNA in enteroids by >100,000-fold, restoring DEFA5 and DEFA6 to levels comparable to those found in primary human tissue. These results newly identify FOXO signaling as a pathway of biological and potentially therapeutic relevance for the regulation of human Paneth cell α-defensins in health and disease.


Asunto(s)
Antiinfecciosos , alfa-Defensinas , Humanos , Animales , Ratones , alfa-Defensinas/genética , alfa-Defensinas/farmacología , alfa-Defensinas/metabolismo , Intestinos , Intestino Delgado/metabolismo , Células de Paneth/metabolismo , Antiinfecciosos/metabolismo , Organoides/metabolismo
4.
PLoS Pathog ; 19(2): e1011200, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36821624

RESUMEN

In the mammalian intestine, flagellar motility can provide microbes competitive advantage, but also threatens the spatial segregation established by the host at the epithelial surface. Unlike microbicidal defensins, previous studies indicated that the protective activities of human α-defensin 6 (HD6), a peptide secreted by Paneth cells of the small intestine, resides in its remarkable ability to bind microbial surface proteins and self-assemble into protective fibers and nets. Given its ability to bind flagellin, we proposed that HD6 might be an effective inhibitor of bacterial motility. Here, we utilized advanced automated live cell fluorescence imaging to assess the effects of HD6 on actively swimming Salmonella enterica in real time. We found that HD6 was able to effectively restrict flagellar motility of individual bacteria. Flagellin-specific antibody, a classic inhibitor of flagellar motility that utilizes a mechanism of agglutination, lost its activity at low bacterial densities, whereas HD6 activity was not diminished. A single amino acid variant of HD6 that was able to bind flagellin, but not self-assemble, lost ability to inhibit flagellar motility. Together, these results suggest a specialized role of HD6 self-assembly into polymers in targeting and restricting flagellar motility.


Asunto(s)
Antiinfecciosos , Células de Paneth , Animales , Humanos , Células de Paneth/metabolismo , Flagelina/metabolismo , Antiinfecciosos/metabolismo , Bacterias/metabolismo , Flagelos/metabolismo , Mamíferos
5.
FASEB J ; 36(3): e22200, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35182405

RESUMEN

Intelectins (intestinal lectins) are highly conserved across chordate evolution and have been implicated in various human diseases, including Crohn's disease (CD). The human genome encodes two intelectin genes, intelectin-1 (ITLN1) and intelectin-2 (ITLN2). Other than its high sequence similarity with ITLN1, little is known about ITLN2. To address this void in knowledge, we report that ITLN2 exhibits discrete, yet notable differences from ITLN1 in primary structure, including a unique amino terminus, as well as changes in amino acid residues associated with the glycan-binding activity of ITLN1. We identified that ITLN2 is a highly abundant Paneth cell-specific product, which localizes to secretory granules, and is expressed as a multimeric protein in the small intestine. In surgical specimens of ileal CD, ITLN2 mRNA levels were reduced approximately five-fold compared to control specimens. The ileal expression of ITLN2 was unaffected by previously reported disease-associated variants in ITLN2 and CD-associated variants in neighboring ITLN1 as well as NOD2 and ATG16L1. ITLN2 mRNA expression was undetectable in control colon tissue; however, in both ulcerative colitis (UC) and colonic CD, metaplastic Paneth cells were found to express ITLN2. Together, the data reported establish the groundwork for understanding ITLN2 function(s) in the intestine, including its possible role in CD.


Asunto(s)
Enfermedad de Crohn/metabolismo , Lectinas/metabolismo , Células de Paneth/metabolismo , Vesículas Secretoras/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Humanos , Lectinas/genética , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
Nat Immunol ; 11(1): 76-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19855381

RESUMEN

Antimicrobial peptides are important effectors of innate immunity throughout the plant and animal kingdoms. In the mammalian small intestine, Paneth cell alpha-defensins are antimicrobial peptides that contribute to host defense against enteric pathogens. To determine if alpha-defensins also govern intestinal microbial ecology, we analyzed the intestinal microbiota of mice expressing a human alpha-defensin gene (DEFA5) and in mice lacking an enzyme required for the processing of mouse alpha-defensins. In these complementary models, we detected significant alpha-defensin-dependent changes in microbiota composition, but not in total bacterial numbers. Furthermore, DEFA5-expressing mice had striking losses of segmented filamentous bacteria and fewer interleukin 17 (IL-17)-producing lamina propria T cells. Our data ascribe a new homeostatic role to alpha-defensins in regulating the makeup of the commensal microbiota.


Asunto(s)
Ecología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , alfa-Defensinas/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Recuento de Colonia Microbiana , Femenino , Citometría de Flujo , Humanos , Hibridación Fluorescente in Situ , Interleucina-17/inmunología , Interleucina-17/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestinos/inmunología , Masculino , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Metagenoma , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Filogenia , ARN Ribosómico 16S/genética , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , alfa-Defensinas/genética , alfa-Defensinas/inmunología
7.
PLoS Pathog ; 15(4): e1007719, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30973939

RESUMEN

The regulation of mucosal immune function is critical to host protection from enteric pathogens but is incompletely understood. The nervous system and the neurotransmitter acetylcholine play an integral part in host defense against enteric bacterial pathogens. Here we report that acetylcholine producing-T-cells, as a non-neuronal source of ACh, were recruited to the colon during infection with the mouse pathogen Citrobacter rodentium. These ChAT+ T-cells did not exclusively belong to one Th subset and were able to produce IFNγ, IL-17A and IL-22. To interrogate the possible protective effect of acetylcholine released from these cells during enteric infection, T-cells were rendered deficient in their ability to produce acetylcholine through a conditional gene knockout approach. Significantly increased C. rodentium burden was observed in the colon from conditional KO (cKO) compared to WT mice at 10 days post-infection. This increased bacterial burden in cKO mice was associated with increased expression of the cytokines IL-1ß, IL-6, and TNFα, but without significant changes in T-cell and ILC associated IL-17A, IL-22, and IFNγ, or epithelial expression of antimicrobial peptides, compared to WT mice. Despite the increased expression of pro-inflammatory cytokines during C. rodentium infection, inducible nitric oxide synthase (Nos2) expression was significantly reduced in intestinal epithelial cells of ChAT T-cell cKO mice 10 days post-infection. Additionally, a cholinergic agonist enhanced IFNγ-induced Nos2 expression in intestinal epithelial cell in vitro. These findings demonstrated that acetylcholine, produced by specialized T-cells that are recruited during C. rodentium infection, are a key mediator in host-microbe interactions and mucosal defenses.


Asunto(s)
Acetilcolina/metabolismo , Citrobacter rodentium/inmunología , Colon/inmunología , Infecciones por Enterobacteriaceae/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Colon/metabolismo , Citocinas/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Interleucina-17/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CXCR5/fisiología
9.
Semin Immunol ; 25(5): 334-41, 2013 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-24239045

RESUMEN

The complex community of colonizing microbes inhabiting the mucosal surfaces of mammals is vital to homeostasis and normal physiology in the host. When the composition of this microbiota is unfavorably altered, termed dysbiosis, the host is rendered more susceptible to a variety of chronic diseases. In the mammalian small intestine, specialized secretory epithelial cells, named Paneth cells, produce a variety of secreted antimicrobial peptides that fundamentally influence the composition of the microbiota. Recent investigations have identified numerous genetic and environmental factors that can disrupt normal Paneth cell function, resulting in compromised antimicrobial peptide secretion and consequent dysbiosis. These findings suggest that Paneth cell dysfunction should be considered a common cause of dysbiosis.


Asunto(s)
Disbiosis/patología , Células de Paneth/fisiología , Animales , Disbiosis/microbiología , Humanos , Microbiota , Células de Paneth/microbiología , Células de Paneth/patología
10.
Annu Rev Physiol ; 75: 289-311, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23398152

RESUMEN

Paneth cells are highly specialized epithelial cells of the small intestine, where they coordinate many physiological functions. First identified more than a century ago on the basis of their readily discernible secretory granules by routine histology, these cells are located at the base of the crypts of Lieberkühn, tiny invaginations that line the mucosal surface all along the small intestine. Investigations over the past several decades determined that these cells synthesize and secrete substantial quantities of antimicrobial peptides and proteins. More recent studies have determined that these antimicrobial molecules are key mediators of host-microbe interactions, including homeostatic balance with colonizing microbiota and innate immune protection from enteric pathogens. Perhaps more intriguing, Paneth cells secrete factors that help sustain and modulate the epithelial stem and progenitor cells that cohabitate in the crypts and rejuvenate the small intestinal epithelium. Dysfunction of Paneth cell biology contributes to the pathogenesis of chronic inflammatory bowel disease.


Asunto(s)
Mucosa Intestinal/fisiología , Intestino Delgado/fisiología , Células de Paneth/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/fisiología , Humanos , Inmunidad Innata/fisiología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/fisiopatología , Metagenoma/fisiología , Ratones
11.
Nature ; 467(7314): 426-9, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20864996

RESUMEN

Salmonella enterica serotype Typhimurium (S. Typhimurium) causes acute gut inflammation by using its virulence factors to invade the intestinal epithelium and survive in mucosal macrophages. The inflammatory response enhances the transmission success of S. Typhimurium by promoting its outgrowth in the gut lumen through unknown mechanisms. Here we show that reactive oxygen species generated during inflammation react with endogenous, luminal sulphur compounds (thiosulphate) to form a new respiratory electron acceptor, tetrathionate. The genes conferring the ability to use tetrathionate as an electron acceptor produce a growth advantage for S. Typhimurium over the competing microbiota in the lumen of the inflamed gut. We conclude that S. Typhimurium virulence factors induce host-driven production of a new electron acceptor that allows the pathogen to use respiration to compete with fermenting gut microbes. Thus the ability to trigger intestinal inflammation is crucial for the biology of this diarrhoeal pathogen.


Asunto(s)
Respiración de la Célula , Electrones , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Salmonella typhimurium/metabolismo , Animales , Colitis/metabolismo , Colitis/microbiología , Transporte de Electrón , Femenino , Tracto Gastrointestinal/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Ácido Tetratiónico/metabolismo , Tiosulfatos/metabolismo
12.
PLoS Pathog ; 9(4): e1003267, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637594

RESUMEN

Chemotaxis enhances the fitness of Salmonella enterica serotype Typhimurium (S. Typhimurium) during colitis. However, the chemotaxis receptors conferring this fitness advantage and their cognate signals generated during inflammation remain unknown. Here we identify respiratory electron acceptors that are generated in the intestinal lumen as by-products of the host inflammatory response as in vivo signals for methyl-accepting chemotaxis proteins (MCPs). Three MCPs, including Trg, Tsr and Aer, enhanced the fitness of S. Typhimurium in a mouse colitis model. Aer mediated chemotaxis towards electron acceptors (energy taxis) in vitro and required tetrathionate respiration to confer a fitness advantage in vivo. Tsr mediated energy taxis towards nitrate but not towards tetrathionate in vitro and required nitrate respiration to confer a fitness advantage in vivo. These data suggest that the energy taxis receptors Tsr and Aer respond to distinct in vivo signals to confer a fitness advantage upon S. Typhimurium during inflammation by enabling this facultative anaerobic pathogen to seek out favorable spatial niches containing host-derived electron acceptors that boost its luminal growth.


Asunto(s)
Proteínas Bacterianas/metabolismo , Quimiotaxis , Colitis/microbiología , Metabolismo Energético , Proteínas de la Membrana/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/patogenicidad , Animales , Proteínas Portadoras/metabolismo , Colitis/inmunología , Transporte de Electrón , Femenino , Inflamación , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Proteínas Quimiotácticas Aceptoras de Metilo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Neutrófilos/inmunología , Nitratos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Superficie Celular/metabolismo , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/fisiología , Ácido Tetratiónico/metabolismo
13.
Pediatr Res ; 76(4): 326-33, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25000347

RESUMEN

BACKGROUND: Probiotics decrease the risk of necrotizing enterocolitis (NEC). We sought to determine the impact of Bifidobacterium longum subsp. infantis (B. infantis) in the established rat model of NEC. METHODS: Rat pups delivered 1 d prior to term gestation were assigned to one of three groups: dam fed (DF), formula fed (FF), or fed with formula supplemented with 5 × 10(6) CFU B. infantis per day (FF+Binf). Experimental pups were exposed to hypoxia and cold stress. Ileal tissue was examined for pathology and expression of inflammatory mediators, antimicrobial peptides, and goblet-cell products. Ceca were assessed for bacterial composition by analysis of the 16S rRNA sequence. RESULTS: Administration of B. infantis significantly reduced the incidence of NEC, decreased expression of Il6, Cxcl1, Tnfa, Il23, and iNOS, and decreased expression of the antimicrobial peptides Reg3b and Reg3g. There was significant microbial heterogeneity both within groups and between experiments. The cecal microbiota was not significantly different between the FF and FF+Binf groups. Bifidobacteria were not detected in the cecum in significant numbers. CONCLUSION: In the rat model, the inflammation associated with NEC was attenuated by administration of probiotic B. infantis. Dysbiosis was highly variable, precluding determination of the precise role of the microbiota in experimental NEC.


Asunto(s)
Bifidobacterium/fisiología , Enterocolitis Necrotizante/terapia , Inmunidad Innata/fisiología , Inflamación/terapia , Microbiota , Animales , Bifidobacterium/genética , Ciego/microbiología , ARN Ribosómico 16S/genética , Ratas
14.
J Immunol Methods ; 525: 113599, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38081407

RESUMEN

Intestinal transplantation is the definitive treatment for intestinal failure. However, tissue rejection and graft-versus-host disease are relatively common complications, necessitating aggressive immunosuppression that can itself pose further complications. Tracking intraluminal markers in ileal effluent from standard ileostomies may present a noninvasive and sensitive way to detect developing pathology within the intestinal graft. This would be an improvement compared to current assessments, which are limited by poor sensitivity and specificity, contributing to under or over-immunosuppression, respectively, and by the need for invasive biopsies. Herein, we report an approach to reproducibly analyze ileal fluid obtained through stoma sampling for antimicrobial peptide/protein concentrations, reasoning that these molecules may provide an assessment of intestinal homeostasis and levels of intestinal inflammation over time. Concentrations of lysozyme (LYZ), myeloperoxidase (MPO), calprotectin (S100A8/A9) and ß-defensin 2 (DEFB2) were assessed using adaptations of commercially available enzyme-linked immunosorbent assays (ELISAs). The concentration of α-defensin 5 (DEFA5) was assessed using a newly developed sandwich ELISA. Our data support that with proper preparation of ileal effluent specimens, precise and replicable determination of antimicrobial peptide/protein concentrations can be achieved for each of these target molecules via ELISA. This approach may prove to be reliable as a clinically useful assessment of intestinal homeostasis over time for patients with ileostomies.


Asunto(s)
Péptidos Antimicrobianos , alfa-Defensinas , Humanos , Intestinos , Ensayo de Inmunoadsorción Enzimática , Biopsia
15.
J Proteome Res ; 12(5): 2295-304, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23586814

RESUMEN

Milk is traditionally considered an ideal source of the basic elemental nutrients required by infants. More detailed examination is revealing that milk represents a more functional ensemble of components with benefits to both infants and mothers. A comprehensive peptidomics method was developed and used to analyze human milk yielding an extensive array of protein products present in the fluid. Over 300 milk peptides were identified originating from major and many minor protein components of milk. As expected, the majority of peptides derived from ß-casein, however no peptide fragments from the major milk proteins lactoferrin, α-lactalbumin, and secretory immunoglobulin A were identified. Proteolysis in the mammary gland is selective-released peptides were drawn only from specific proteins and typically from only select parts of the parent sequence. A large number of the peptides showed significant sequence overlap with peptides with known antimicrobial or immunomodulatory functions. Antibacterial assays showed the milk peptide mixtures inhibited the growth of Escherichia coli and Staphylococcus aureus . The predigestion of milk proteins and the consequent release of antibacterial peptides may provide a selective advantage through evolution by protecting both the mother's mammary gland and her nursing offspring from infection.


Asunto(s)
Antibacterianos/química , Proteínas de la Leche/química , Leche Humana/química , Fragmentos de Péptidos/química , Secuencia de Aminoácidos , Antibacterianos/farmacología , Pruebas Antimicrobianas de Difusión por Disco , Escherichia coli/efectos de los fármacos , Femenino , Humanos , Proteínas de la Leche/farmacología , Datos de Secuencia Molecular , Fragmentos de Péptidos/farmacología , Proteolisis , Proteómica , Staphylococcus aureus/efectos de los fármacos
16.
Hepatology ; 55(4): 1154-63, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22095436

RESUMEN

UNLABELLED: Liver cirrhosis is associated with bacterial translocation (BT) and endotoxemia. Most translocating bacteria belong to the common intestinal microbiota, suggesting a breakdown of intestinal barrier function. We hypothesized that diminished mucosal antimicrobial host defense could predispose to BT. Two rodent models of portal hypertension with increased BT were used, CCl(4)-induced ascitic cirrhosis and 2-day portal vein-ligated (PVL) animals. BT was assessed by standard microbiological techniques on mesenteric lymph nodes. Total RNA was isolated systematically throughout the intestinal tract, and expression of Paneth cell α-cryptdins and ß-defensins was determined by real-time quantitative polymerase chain reaction (qPCR). To determine functional consequences, mucosal antimicrobial activity was assessed with a fluorescence-activated cell sorting assay. BT was detectable in 40% of rats with cirrhosis. Compared with the group without BT, these animals exhibited diminished intestinal Paneth cell α-cryptdin 5 and 7 expression. In contrast, PVL was associated with BT in all animals but did not affect antimicrobial peptides. The decrease in Paneth cell antimicrobials was most pronounced in the ileum and the coecum. Other antimicrobials showed no changes or even an induction in the case of BT at different sites. Antimicrobial activity toward different commensal strains was reduced, especially in the distal ileum and the cecum in experimental cirrhosis with BT (excluding PVL). CONCLUSION: Compromised Paneth cell antimicrobial host defense seems to predispose to BT in experimental cirrhosis. Understanding this liver-gut axis including the underlying mechanisms could help us to find new treatment avenues.


Asunto(s)
Traslocación Bacteriana/fisiología , Intestinos/microbiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Células de Paneth/metabolismo , Precursores de Proteínas/metabolismo , beta-Defensinas/metabolismo , Animales , Bacteroides fragilis/fisiología , Bifidobacterium/fisiología , Tetracloruro de Carbono/efectos adversos , Ciego/microbiología , Modelos Animales de Enfermedad , Enterococcus faecalis/fisiología , Escherichia coli/fisiología , Hipertensión Portal/etiología , Hipertensión Portal/fisiopatología , Íleon/microbiología , Ligadura/efectos adversos , Cirrosis Hepática/inducido químicamente , Masculino , Células de Paneth/patología , Vena Porta/fisiopatología , Ratas , Ratas Endogámicas
17.
Dig Dis ; 31(3-4): 299-304, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24246978

RESUMEN

The intestinal mucosa interfaces with a complex, dense community of microorganisms, including hundreds of species of resident microbiota and many transient microbes entering from food- and water-borne sources. In the small intestine, Paneth cells (specialized secretory epithelial cells) produce abundant quantities of α-defensins and several other antibiotic peptides. Human Paneth cells make two α-defensins: HD5 and HD6. Data from in vivo models indicate that Paneth cell α-defensins play a pivotal role in defense from food- and water-borne pathogens in the intestine. The mechanism by which these two α-defensins protect from enteric pathogens is quite distinct. HD5 is a potent antimicrobial that kills target microbes by membrane disruption, whereas HD6 is newly discovered to self-assemble to form fibrils and nanonets that surround and entangle bacteria. Recent data suggest that HD5 also serves to help shape the composition of the colonizing microbiota. Studies in humans suggest that reduced expression of HD5 and HD6 is a fundamental feature of ileal Crohn's disease. Mechanistically, the link between reduced Paneth cell α-defensin expression and ileal Crohn's disease pathogenesis may be a result of the weakened mucosal antimicrobial defense and/or alterations in the composition of commensal microbiota.


Asunto(s)
Inmunidad Innata , Intestino Delgado/inmunología , alfa-Defensinas/inmunología , Animales , Humanos , Mucosa Intestinal/inmunología , Modelos Biológicos , Células de Paneth/inmunología
18.
Proc Natl Acad Sci U S A ; 107(33): 14739-44, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20679225

RESUMEN

Mutations in the NOD2 gene are strong genetic risk factors for ileal Crohn's disease. However, the mechanism by which these mutations predispose to intestinal inflammation remains a subject of controversy. We report that Nod2-deficient mice inoculated with Helicobacter hepaticus, an opportunistic pathogenic bacterium, developed granulomatous inflammation of the ileum, characterized by an increased expression of Th1-related genes and inflammatory cytokines. The Peyer's patches and mesenteric lymph nodes were markedly enlarged with expansion of IFN-gamma-producing CD4 and CD8 T cells. Rip2-deficient mice exhibited a similar phenotype, suggesting that Nod2 function likely depends on the Rip2 kinase in this model. Transferring wild-type bone marrow cells into irradiated Nod2-deficient mice did not rescue the phenotype. However, restoring crypt antimicrobial function of Nod2-deficient mice by transgenic expression of alpha-defensin in Paneth cells rescued the Th1 inflammatory phenotype. Therefore, through the regulation of intestinal microbes, Nod2 function in nonhematopoietic cells of the small intestinal crypts is critical for protecting mice from a Th1-driven granulomatous inflammation in the ileum. The model may provide insight into Nod2 function relevant to inflammation of ileal Crohn's disease.


Asunto(s)
Enfermedad de Crohn/inmunología , Infecciones por Helicobacter/inmunología , Íleon/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Animales , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Femenino , Citometría de Flujo , Infecciones por Helicobacter/microbiología , Helicobacter hepaticus/inmunología , Helicobacter hepaticus/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Íleon/metabolismo , Íleon/microbiología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células TH1/inmunología , Células TH1/metabolismo
19.
Nat Biomed Eng ; 7(1): 38-55, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36550307

RESUMEN

The microbiome modulates host immunity and aids the maintenance of tolerance in the gut, where microbial and food-derived antigens are abundant. Yet modern dietary factors and the excessive use of antibiotics have contributed to the rising incidence of food allergies, inflammatory bowel disease and other non-communicable chronic diseases associated with the depletion of beneficial taxa, including butyrate-producing Clostridia. Here we show that intragastrically delivered neutral and negatively charged polymeric micelles releasing butyrate in different regions of the intestinal tract restore barrier-protective responses in mouse models of colitis and of peanut allergy. Treatment with the butyrate-releasing micelles increased the abundance of butyrate-producing taxa in Clostridium cluster XIVa, protected mice from an anaphylactic reaction to a peanut challenge and reduced disease severity in a T-cell-transfer model of colitis. By restoring microbial and mucosal homoeostasis, butyrate-releasing micelles may function as an antigen-agnostic approach for the treatment of allergic and inflammatory diseases.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Hipersensibilidad al Cacahuete , Ratones , Animales , Micelas , Butiratos
20.
Pediatr Res ; 71(5): 546-51, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22322385

RESUMEN

INTRODUCTION: Necrotizing enterocolitis (NEC) is a devastating disease of premature infants. Probiotics decrease the risk of NEC in clinical and experimental studies. Antimicrobial peptides protect the gut against noxious microbes and shape the commensal microbiota, but their role in NEC remains unclear. METHODS: To investigate the expression of antimicrobial peptides in experimental NEC and the impact of probiotics on their expression, premature rats were divided into three groups: dam fed (DF), hand fed with formula (FF), or hand fed with formula containing Bifidobacterium bifidum (FF + BIF). All groups were exposed to asphyxia and cold stress. RESULTS: Like in human ontogeny, the rat pup has low expression of Paneth cell antimicrobials, which increases rapidly during normal development. The expression of lysozyme, secretory phospholipase A(2) (sPLA(2)), pancreatic-associated proteins 1 and 3 mRNA was elevated in the FF group with a high incidence of NEC, as compared with the DF and FF + BIF groups where the disease was attenuated. DISCUSSION: We conclude that induction of antimicrobial peptides occurs in experimental NEC similar to that reported in human disease and is attenuated when disease is averted by probiotic B. bifidum. The induction of antimicrobial peptides is likely an adaptive mucosal response that is often not sufficient to prevent disease in the premature gut.


Asunto(s)
Bifidobacterium , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/microbiología , Péptidos/metabolismo , Probióticos , Proteínas/metabolismo , Animales , Animales Recién Nacidos , Antiinfecciosos , Secuencia de Bases , Cartilla de ADN , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/terapia , Expresión Génica , Inmunohistoquímica , Péptidos/genética , Reacción en Cadena de la Polimerasa , Proteínas/genética , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA