Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Can Vet J ; 65(9): 886-893, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39219609

RESUMEN

A 15-month-old spayed female greater Swiss mountain dog was brought to our clinic because of relapsing episodes of urinary tract infection, present since her adoption at 2 mo of age. A diagnosis of chronic bacterial cystitis associated with an invasive, biofilm-forming uropathogenic Escherichia coli was made with bladder-wall histology and fluorescent in situ hybridization analysis. Local treatment with EDTA-tromethamine (EDTA-Tris) infusions along with parenteral cefquinome and prophylactic measures (Type-A proanthocyanidins and probiotics) coincided with clinical and bacterial remission. The dog has been free of clinical signs of urinary tract infection for >4 y. Biofilm-forming uropathogenic E. coli can cause chronic, recurrent cystitis due to low antibiotic efficacy and should be considered in cases of recurrent cystitis in dogs, especially in the absence of identified predisposing factors. This case report describes the diagnostic and therapeutic options that were used to manage a case of this type. Key clinical message: Fluorescent in situ hybridization analysis may be considered in the diagnosis of chronic bacterial cystitis in dogs, and intravesical instillations of EDTA-Tris may be helpful in managing such cases.


Traitement adjuvant intravésical avec de l'EDTA-trométhamine chez un chien présentant une cystite récurrente à Escherichia coli formant des biofilmsUne chienne grand bouvier suisse stérilisée de 15 mois nous a été présentée pour des épisodes d'infection du tractus urinaire récidivants depuis son adoption à l'âge de 2 mois. Une cystite bactérienne chronique associée à un Escherichia coli uropathogène formant des biofilms a été identifiée par l'examen histologique de la paroi vésicale et par hybridation in situ fluorescente. Des instillations intravésicales d'EDTA et trométhamine (EDTA-Tris) en complément d'une antibiothérapie parentérale de courte durée (cefquinome) et de mesures prophylactiques (proanthocyanidines de type A et probiotiques) ont permis une guérison clinique et bactériologique de la cystite pendant plus de 4 ans. Les infections par Escherichia coli formant des biofilms peuvent causer des cystites chroniques récurrentes dues à une faible efficacité des antibiotiques et doivent être incluses dans le diagnostic différentiel des cystites récurrentes chez le chien, particulièrement en l'absence d'autre facteur prédisposant. Ce rapport propose des stratégies diagnostiques et thérapeutiques ayant permis la prise en charge d'un de ces cas.Message clinique clé :L'analyse par hybridation in situ fluorescente peut être envisagé dans le diagnostic de cystite bactérienne chronique chez les chiens, et l'instillation intravésicale d'EDTA-Tris peut être utile dans la gestion de tels cas.(Traduit par les auteurs).


Asunto(s)
Antibacterianos , Biopelículas , Cistitis , Enfermedades de los Perros , Ácido Edético , Infecciones por Escherichia coli , Perros , Animales , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/microbiología , Femenino , Cistitis/veterinaria , Cistitis/tratamiento farmacológico , Cistitis/microbiología , Ácido Edético/uso terapéutico , Ácido Edético/administración & dosificación , Biopelículas/efectos de los fármacos , Infecciones por Escherichia coli/veterinaria , Infecciones por Escherichia coli/tratamiento farmacológico , Antibacterianos/uso terapéutico , Antibacterianos/administración & dosificación , Administración Intravesical , Escherichia coli/efectos de los fármacos , Recurrencia
2.
Br J Clin Pharmacol ; 90(1): 107-126, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37559444

RESUMEN

AIMS: Several medicinal treatments for avoiding postoperative ileus (POI) after abdominal surgery have been evaluated in randomized controlled trials (RCTs). This network meta-analysis aimed to explore the relative effectiveness of these different treatments on ileus outcome measures. METHODS: A systematic literature review was performed to identify RCTs comparing treatments for POI following abdominal surgery. A Bayesian network meta-analysis was performed. Direct and indirect comparisons of all regimens were simultaneously compared using random-effects network meta-analysis. RESULTS: A total of 38 RCTs were included in this network meta-analysis reporting on 6371 patients. Our network meta-analysis shows that prokinetics significantly reduce the duration of first gas (mean difference [MD] = 16 h; credible interval -30, -3.1; surface under the cumulative ranking curve [SUCRA] 0.418), duration of first bowel movements (MD = 25 h; credible interval -39, -11; SUCRA 0.25) and duration of postoperative hospitalization (MD -1.9 h; credible interval -3.8, -0.040; SUCRA 0.34). Opioid antagonists are the only treatment that significantly improve the duration of food recovery (MD -19 h; credible interval -26, -14; SUCRA 0.163). CONCLUSION: Based on our meta-analysis, the 2 most consistent pharmacological treatments able to effectively reduce POI after abdominal surgery are prokinetics and opioid antagonists. The absence of clear superiority of 1 treatment over another highlights the limits of the pharmacological principles available.


Asunto(s)
Ileus , Antagonistas de Narcóticos , Humanos , Metaanálisis en Red , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/prevención & control , Ileus/tratamiento farmacológico , Ileus/etiología , Ileus/prevención & control
3.
Commun Biol ; 6(1): 1168, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37968381

RESUMEN

Opioid-dependent immune-mediated analgesic effects have been broadly reported upon inflammation. In preclinical mouse models of intestinal inflammatory diseases, the local release of enkephalins (endogenous opioids) by colitogenic T lymphocytes alleviate inflammation-induced pain by down-modulating gut-innervating nociceptor activation in periphery. In this study, we wondered whether this immune cell-derived enkephalin-mediated regulation of the nociceptor activity also operates under steady state conditions. Here, we show that chimeric mice engrafted with enkephalin-deficient bone marrow cells exhibit not only visceral hypersensitivity but also an increase in both epithelial paracellular and transcellular permeability, an alteration of the microbial topography resulting in increased bacteria-epithelium interactions and a higher frequency of IgA-producing plasma cells in Peyer's patches. All these alterations of the intestinal homeostasis are associated with an anxiety-like behavior despite the absence of an overt inflammation as observed in patients with irritable bowel syndrome. Thus, our results show that immune cell-derived enkephalins play a pivotal role in maintaining gut homeostasis and normal behavior in mice. Because a defect in the mucosal opioid system remarkably mimics some major clinical symptoms of the irritable bowel syndrome, its identification might help to stratify subgroups of patients.


Asunto(s)
Síndrome del Colon Irritable , Humanos , Animales , Ratones , Analgésicos Opioides , Encefalinas/genética , Inflamación , Dolor
4.
Antioxid Redox Signal ; 36(4-6): 211-219, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33691464

RESUMEN

Significance: Hydrogen sulfide (H2S), an important regulator of physiology and health, helps resolve inflammation and promotes tissue repair in the gastrointestinal tract. Recent Advances: Gut microbiota live as a multispecies biofilm in close interaction with the upper mucus layer lining the epithelium. The relative abundance, spatial organization, and function of these microorganisms affect a broad range of health outcomes. This article provides a state-of-the-art review of our understanding of the cross talk between H2S, the gut microbiota, and health. H2S can have toxic or therapeutic effects, depending on its concentration and source. When produced at excessive concentrations by local microbiota, H2S may cause mucus disruption and inflammation and contribute to development of cancer. In contrast, low levels of endogenous or exogenous H2S directly stabilize mucus layers, prevent fragmentation and adherence of the microbiota biofilm to the epithelium, inhibit the release of invasive pathobionts, and help resolve inflammation and tissue injury. Although scarce, research findings suggest that dietary H2S obtained from plants or ingestion of the H2S precursor, L-cysteine, may also modulate the abundance and function of microbiota. Critical Issues: A critical issue is the lack of understanding of the metagenomic, transcriptomic, and proteomic alterations that characterize the interactions between H2S and gut microbiota to shape health outcomes. Future Directions: The ambivalent roles of H2S in the gut offer a fertile ground for research on such critical issues. The findings will improve our understanding of how H2S modulates the microbiota to affect body function and will help identify novel therapeutic strategies. Antioxid. Redox Signal. 36, 211-219.


Asunto(s)
Microbioma Gastrointestinal , Sulfuro de Hidrógeno , Microbiota , Tracto Gastrointestinal , Sulfuro de Hidrógeno/farmacología , Proteómica
5.
Inflamm Bowel Dis ; 27(Suppl 2): S33-S37, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34791291

RESUMEN

A new paradigm has been added for the treatment of inflammatory bowel diseases such as Crohn's disease and ulcerative colitis. In addition to resolving symptoms and inflammatory cell activation, the objective of tissue repair and mucosal healing is also now considered a primary goal. In the search of mediators that would be responsible for delayed mucosal healing, protease-activated receptor-1 (PAR-1) has emerged as a most interesting target. Indeed, in Crohn's disease, the endogenous PAR-1 agonist thrombin is drastically activated. Activation of PAR-1 is known to be associated with epithelial dysfunctions that hamper mucosal homeostasis. This review gathers the scientific evidences of a potential role for PAR-1 in mucosal damage and mucosal dysfunctions associated with chronic intestinal inflammation. The potential clinical benefits of PAR-1 antagonism to promote mucosal repair in CD patients are discussed. Targeted local delivery of a PAR-1 antagonist molecule such as CVT120165, a formulated version of the FDA-approved PAR-1 antagonist vorapaxar, at the mucosa of Crohn's disease patients could be proposed as a new indication for IBD that could be rapidly tested in clinical trials.


The potential clinical benefits and indications of PAR-1 antagonism to treat inflammatory bowel diseases are discussed.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Receptor PAR-1/uso terapéutico , Trombina , Colitis Ulcerosa , Humanos , Enfermedades Inflamatorias del Intestino , Receptor PAR-1/genética
6.
mSphere ; 6(4): e0062421, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34378987

RESUMEN

The probiotic Escherichia coli strain Nissle 1917 (DSM 6601, Mutaflor), generally considered beneficial and safe, has been used for a century to treat various intestinal diseases. However, Nissle 1917 hosts in its genome the pks pathogenicity island that codes for the biosynthesis of the genotoxin colibactin. Colibactin is a potent DNA alkylator, suspected to play a role in colorectal cancer development. We show in this study that Nissle 1917 is functionally capable of producing colibactin and inducing interstrand cross-links in the genomic DNA of epithelial cells exposed to the probiotic. This toxicity was even exacerbated with lower doses of the probiotic, when the exposed cells started to divide again but exhibited aberrant anaphases and increased gene mutation frequency. DNA damage was confirmed in vivo in mouse models of intestinal colonization, demonstrating that Nissle 1917 produces the genotoxin in the gut lumen. Although it is possible that daily treatment of adult humans with their microbiota does not produce the same effects, administration of Nissle 1917 as a probiotic or as a chassis to deliver therapeutics might exert long-term adverse effects and thus should be considered in a risk-versus-benefit evaluation. IMPORTANCE Nissle 1917 is sold as a probiotic and considered safe even though it has been known since 2006 that it harbors the genes for colibactin synthesis. Colibactin is a potent genotoxin that is now linked to causative mutations found in human colorectal cancer. Many papers concerning the use of this strain in clinical applications ignore or elude this fact or misleadingly suggest that Nissle 1917 does not induce DNA damage. Here, we demonstrate that Nissle 1917 produces colibactin in vitro and in vivo and induces mutagenic DNA damage. This is a serious safety concern that must not be ignored in the interests of patients, the general public, health care professionals, and ethical probiotic manufacturers.


Asunto(s)
Daño del ADN , Células Epiteliales/microbiología , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Genoma Bacteriano , Mutagénesis , Probióticos , Animales , Células CHO , Cricetulus , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Femenino , Islas Genómicas , Células HeLa , Humanos , Ratones , Mutación
7.
PLoS Pathog ; 17(2): e1009310, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33630958

RESUMEN

Urinary tract infections (UTIs) are among the most common outpatient infections, with a lifetime incidence of around 60% in women. We analysed urine samples from 223 patients with community-acquired UTIs and report the presence of the cleavage product released during the synthesis of colibactin, a bacterial genotoxin, in 55 of the samples examined. Uropathogenic Escherichia coli strains isolated from these patients, as well as the archetypal E. coli strain UTI89, were found to produce colibactin. In a murine model of UTI, the machinery producing colibactin was expressed during the early hours of the infection, when intracellular bacterial communities form. We observed extensive DNA damage both in umbrella and bladder progenitor cells. To the best of our knowledge this is the first report of colibactin production in UTIs in humans and its genotoxicity in bladder cells.


Asunto(s)
Daño del ADN , Infecciones por Escherichia coli/patología , Péptidos/metabolismo , Policétidos/metabolismo , Vejiga Urinaria/patología , Infecciones Urinarias/patología , Escherichia coli Uropatógena/aislamiento & purificación , Anciano , Animales , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Mutágenos/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/microbiología , Infecciones Urinarias/genética , Infecciones Urinarias/microbiología
8.
J Crohns Colitis ; 15(5): 787-799, 2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-33201214

RESUMEN

BACKGROUND AND AIMS: Thrombin levels in the colon of Crohn's disease patients have recently been found to be elevated 100-fold compared with healthy controls. Our aim was to determine whether and how dysregulated thrombin activity could contribute to local tissue malfunctions associated with Crohn's disease. METHODS: Thrombin activity was studied in tissues from Crohn's disease patients and healthy controls. Intracolonic administration of thrombin to wild-type or protease-activated receptor-deficient mice was used to assess the effects and mechanisms of local thrombin upregulation. Colitis was induced in rats and mice by the intracolonic administration of trinitrobenzene sulphonic acid. RESULTS: Active forms of thrombin were increased in Crohn's disease patient tissues. Elevated thrombin expression and activity were associated with intestinal epithelial cells. Increased thrombin activity and expression were also a feature of experimental colitis in rats. Colonic exposure to doses of active thrombin comparable to what is found in inflammatory bowel disease tissues caused mucosal damage and tissue dysfunctions in mice, through a mechanism involving both protease-activated receptors -1 and -4. Intracolonic administration of the thrombin inhibitor dabigatran, as well as inhibition of protease-activated receptor-1, prevented trinitrobenzene sulphonic acid-induced colitis in rodent models. CONCLUSIONS: Our data demonstrated that increased local thrombin activity, as it occurs in the colon of patients with inflammatory bowel disease, causes mucosal damage and inflammation. Colonic thrombin and protease-activated receptor-1 appear as possible mechanisms involved in mucosal damage and loss of function and therefore represent potential therapeutic targets for treating inflammatory bowel disease.


Asunto(s)
Enfermedad de Crohn/metabolismo , Receptores Proteinasa-Activados/metabolismo , Trombina/metabolismo , Animales , Estudios de Casos y Controles , Femenino , Humanos , Lactonas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Piridinas/farmacología , Ratas , Ratas Wistar , Regulación hacia Arriba
9.
Nat Commun ; 10(1): 3224, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324782

RESUMEN

Proteolytic homeostasis is important at mucosal surfaces, but its actors and their precise role in physiology are poorly understood. Here we report that healthy human and mouse colon epithelia are a major source of active thrombin. We show that mucosal thrombin is directly regulated by the presence of commensal microbiota. Specific inhibition of luminal thrombin activity causes macroscopic and microscopic damage as well as transcriptomic alterations of genes involved in host-microbiota interactions. Further, luminal thrombin inhibition impairs the spatial segregation of microbiota biofilms, allowing bacteria to invade the mucus layer and to translocate across the epithelium. Thrombin cleaves the biofilm matrix of reconstituted mucosa-associated human microbiota. Our results indicate that thrombin constrains biofilms at the intestinal mucosa. Further work is needed to test whether thrombin plays similar roles in other mucosal surfaces, given that lung, bladder and skin epithelia also express thrombin.


Asunto(s)
Bacterias/metabolismo , Biopelículas , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/microbiología , Trombina/metabolismo , Animales , Línea Celular , Colon/microbiología , Neoplasias del Colon/microbiología , Epitelio/microbiología , Homeostasis , Humanos , Pulmón , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Piel , Trombina/genética , Vejiga Urinaria
10.
J Biomed Sci ; 26(1): 1, 2019 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-30602371

RESUMEN

Gut microbiota interacting with an intact mucosal surface are key to the maintenance of homeostasis and health. This review discusses the current state of knowledge of the biofilm mode of growth of these microbiota communities, and how in turn their disruptions may cause disease. Beyond alterations of relative microbial abundance and diversity, the aim of the review is to focus on the disruptions of the microbiota biofilm structure and function, the dispersion of commensal bacteria, and the mechanisms whereby these dispersed commensals may become pathobionts. Recent findings have linked iron acquisition to the expression of virulence factors in gut commensals that have become pathobionts. Causal studies are emerging, and mechanisms common to enteropathogen-induced disruptions, as well as those reported for Inflammatory Bowel Disease and colo-rectal cancer are used as examples to illustrate the great translational potential of such research. These new observations shed new light on our attempts to develop new therapies that are able to protect and restore gut microbiota homeostasis in the many disease conditions that have been linked to microbiota dysbiosis.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Biopelículas , Disbiosis/fisiopatología , Microbioma Gastrointestinal/fisiología , Enfermedades Inflamatorias del Intestino/inmunología , Hierro/metabolismo , Disbiosis/inmunología , Disbiosis/microbiología , Homeostasis , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/microbiología , Simbiosis
11.
Inflamm Bowel Dis ; 24(7): 1493-1502, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29788224

RESUMEN

Significant alterations of intestinal microbiota and anemia are hallmarks of inflammatory bowel disease (IBD). It is widely accepted that iron is a key nutrient for pathogenic bacteria, but little is known about its impact on microbiota associated with IBD. We used a model device to grow human mucosa-associated microbiota in its physiological anaerobic biofilm phenotype. Compared to microbiota from healthy donors, microbiota from IBD patients generate biofilms ex vivo that were larger in size and cell numbers, contained higher intracellular iron concentrations, and exhibited heightened virulence in a model of human intestinal epithelia in vitro and in the nematode Caenorhabditis elegans. We also describe an unexpected iron-scavenging property for an experimental hydrogen sulfide-releasing derivative of mesalamine. The findings demonstrate that this new drug reduces the virulence of IBD microbiota biofilms through a direct reduction of microbial iron intake and without affecting bacteria survival or species composition within the microbiota. Metabolomic analyses indicate that this drug reduces the intake of purine nucleosides (guanosine), increases the secretion of metabolite markers of purine catabolism (urate and hypoxanthine), and reduces the secretion of uracil (a pyrimidine nucleobase) in complex multispecies human biofilms. These findings demonstrate a new pathogenic mechanism for dysbiotic microbiota in IBD and characterize a novel mode of action for a class of mesalamine derivatives. Together, these observations pave the way towards a new therapeutic strategy for treatment of patients with IBD.


Asunto(s)
Biopelículas , Disbiosis/fisiopatología , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino/microbiología , Hierro/metabolismo , Adulto , Animales , Fenómenos Fisiológicos Bacterianos , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Disbiosis/microbiología , Femenino , Homeostasis , Humanos , Sulfuro de Hidrógeno , Enfermedades Inflamatorias del Intestino/complicaciones , Masculino , Mesalamina/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
12.
Am J Pathol ; 187(11): 2486-2498, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28823873

RESUMEN

The intestinal mucous layer provides a critical host defense against pathogen exposure and epithelial injury, yet little is known about how enteropathogens may circumvent this physiologic barrier. Giardia duodenalis is a small intestinal parasite responsible for diarrheal disease and chronic postinfectious illness. This study reveals a complex interaction at the surface of epithelial cells, between G. duodenalis and the intestinal mucous layer. Here, we reveal mechanisms whereby G. duodenalis evades and disrupts the first line of host defense by degrading human mucin-2 (MUC2), depleting mucin stores and inducing differential gene expression in the mouse small and large intestines. Human colonic biopsy specimens exposed to G. duodenalis were depleted of mucus, and in vivo mice infected with G. duodenalis had a thinner mucous layer and demonstrated differential Muc2 and Muc5ac mucin gene expression. Infection in Muc2-/- mice elevated trophozoite colonization in the small intestine and impaired weight gain. In vitro, human LS174T goblet-like cells were depleted of mucus and had elevated levels of MUC2 mRNA expression after G. duodenalis exposure. Importantly, the cysteine protease inhibitor E64 prevented mucous degradation, mucin depletion, and the increase in MUC2 expression. This article describes a novel role for Giardia's cysteine proteases in pathogenesis and how Giardia's disruptions of the mucous barrier facilitate bacterial translocation that may contribute to the onset and propagation of disease.


Asunto(s)
Células Epiteliales/metabolismo , Giardiasis/genética , Mucinas/genética , Moco/metabolismo , Animales , Traslocación Bacteriana/genética , Proteasas de Cisteína/metabolismo , Femenino , Giardia lamblia/genética , Giardiasis/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Mucinas/metabolismo
13.
PLoS One ; 12(6): e0178647, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28622393

RESUMEN

Our understanding of polymicrobial gastrointestinal infections and their effects on host biology remains incompletely understood. Giardia duodenalis is an ubiquitous intestinal protozoan parasite infecting animals and humans. Concomitant infections with Giardia and other gastrointestinal pathogens commonly occur. In countries with poor sanitation, Giardia infection has been associated with decreased incidence of diarrheal disease and fever, and reduced serum inflammatory markers release, via mechanisms that remain obscure. This study analyzed Giardia spp. co-infections with attaching and effacing (A/E) pathogens, and assessed whether and how the presence of Giardia modulates host responses to A/E enteropathogens, and alters intestinal disease outcome. In mice infected with the A/E pathogen Citrobacter rodentium, co-infection with Giardia muris significantly attenuated weight loss, macro- and microscopic signs of colitis, bacterial colonization and translocation, while concurrently enhancing the production and secretion of antimicrobial peptides (AMPs) mouse ß-defensin 3 and trefoil factor 3 (TFF3). Co-infection of human intestinal epithelial cells (Caco-2) monolayers with G. duodenalis trophozoites and enteropathogenic Escherichia coli (EPEC) enhanced the production of the AMPs human ß-defensin 2 (HBD-2) and TFF3; this effect was inhibited with treatment of G. duodenalis with cysteine protease inhibitors. Collectively, these results suggest that Giardia infections are capable of reducing enteropathogen-induced colitis while increasing production of host AMPs. Additional studies also demonstrated that Giardia was able to directly inhibit the growth of pathogenic bacteria. These results reveal novel mechanisms whereby Giardia may protect against gastrointestinal disease induced by a co-infecting A/E enteropathogen. Our findings shed new light on how microbial-microbial interactions in the gut may protect a host during concomitant infections.


Asunto(s)
Coinfección/metabolismo , Escherichia coli Enteropatógena , Infecciones por Escherichia coli/metabolismo , Giardia lamblia , Giardiasis/metabolismo , Factor Trefoil-3/metabolismo , beta-Defensinas/metabolismo , Animales , Células CACO-2 , Coinfección/microbiología , Coinfección/parasitología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/parasitología , Humanos , Masculino , Ratones
14.
Int J Parasitol ; 47(6): 311-326, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237889

RESUMEN

Giardia duodenalis is a prevalent cause of acute diarrheal disease worldwide. However, recent outbreaks in Italy and Norway have revealed a link between giardiasis and the subsequent development of chronic post-infectious irritable bowel syndrome. While the mechanisms underlying the causation of post-infectious irritable bowel syndrome remain obscure, recent findings suggest that alterations in gut microbiota communities are linked to the pathophysiology of irritable bowel syndrome. In the present study, we use a laboratory biofilm system to culture and enrich mucosal microbiota from human intestinal biopsies. Subsequently, we show that co-culture with Giardia induces disturbances in biofilm species composition and biofilm structure resulting in microbiota communities that are intrinsically dysbiotic - even after the clearance of Giardia. These microbiota abnormalities were mediated in part by secretory-excretory Giardia cysteine proteases. Using in vitro cell culture and germ-free murine infection models, we show that Giardia-induced disruptions of microbiota promote bacterial invasion, resulting in epithelial apoptosis, tight junctional disruption, and bacterial translocation across an intestinal epithelial barrier. Additionally, these dysbiotic microbiota communities resulted in increased activation of the Toll-like receptor 4 signalling pathway, and overproduction of the pro-inflammatory cytokine IL-1beta in humanized germ-free mice. Previous studies that have sought explanations and risk factors for the development of post-infectious irritable bowel syndrome have focused on features of enteropathogens and attributes of the infected host. We propose that polymicrobial interactions involving Giardia and gut microbiota may cause persistent dysbiosis, offering a new interpretation of the reasons why those afflicted with giardiasis are predisposed to gastrointestinal disorders post-infection.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Microbioma Gastrointestinal/fisiología , Giardia lamblia/fisiología , Giardiasis/complicaciones , Síndrome del Colon Irritable/etiología , Animales , Apoptosis , Biopsia , Células CACO-2 , Colon/microbiología , Colon/patología , Proteasas de Cisteína/metabolismo , Heces/microbiología , Heces/parasitología , Vida Libre de Gérmenes , Giardia lamblia/enzimología , Giardia lamblia/ultraestructura , Giardiasis/parasitología , Humanos , Mucosa Intestinal/microbiología , Ratones , Microscopía Electrónica de Rastreo , Conejos , Ratas , Simbiosis
15.
Gut ; 66(10): 1767-1778, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28096305

RESUMEN

OBJECTIVES: Proteases are key mediators of pain and altered enteric neuronal signalling, although the types and sources of these important intestinal mediators are unknown. We hypothesised that intestinal epithelium is a major source of trypsin-like activity in patients with IBS and this activity signals to primary afferent and enteric nerves and induces visceral hypersensitivity. DESIGN: Trypsin-like activity was determined in tissues from patients with IBS and in supernatants of Caco-2 cells stimulated or not. These supernatants were also applied to cultures of primary afferents. mRNA isoforms of trypsin (PRSS1, 2 and 3) were detected by reverse transcription-PCR, and trypsin-3 protein expression was studied by western blot analysis and immunohistochemistry. Electrophysiological recordings and Ca2+ imaging in response to trypsin-3 were performed in mouse primary afferent and in human submucosal neurons, respectively. Visceromotor response to colorectal distension was recorded in mice administered intracolonically with trypsin-3. RESULTS: We showed that stimulated intestinal epithelial cells released trypsin-like activity specifically from the basolateral side. This activity was able to activate sensory neurons. In colons of patients with IBS, increased trypsin-like activity was associated with the epithelium. We identified that trypsin-3 was the only form of trypsin upregulated in stimulated intestinal epithelial cells and in tissues from patients with IBS. Trypsin-3 was able to signal to human submucosal enteric neurons and mouse sensory neurons, and to induce visceral hypersensitivity in vivo, all by a protease-activated receptor-2-dependent mechanism. CONCLUSIONS: In IBS, the intestinal epithelium produces and releases the active protease trypsin-3, which is able to signal to enteric neurons and to induce visceral hypersensitivity.


Asunto(s)
Células Epiteliales/enzimología , Mucosa Intestinal/enzimología , Síndrome del Colon Irritable/enzimología , Síndrome del Colon Irritable/genética , Tripsina/genética , Tripsina/metabolismo , Animales , Células CACO-2 , Estudios de Casos y Controles , Colon/enzimología , Colon/inervación , Medios de Cultivo Condicionados/farmacología , Dipéptidos/farmacología , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/diagnóstico por imagen , Sistema Nervioso Entérico/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Femenino , Ganglios Espinales/citología , Humanos , Hipersensibilidad/enzimología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Isoxazoles/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Microscopía Confocal , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Permeabilidad/efectos de los fármacos , ARN Mensajero/análisis , Ratas , Receptor PAR-2/antagonistas & inhibidores , Receptor PAR-2/metabolismo , Tripsina/farmacología , Tripsinógeno/genética , Regulación hacia Arriba
16.
Inflamm Bowel Dis ; 21(5): 1006-17, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25738373

RESUMEN

BACKGROUND: Microbiota dysbiosis and impaired barrier function are among the most prominent features of inflammatory bowel disease. In the gastrointestinal tract, hydrogen sulfide (H(2)S) is an important regulator of mucosal homeostasis. We hypothesized that H(2)S promotes resolution of colonic inflammation through actions on microbiota biofilm and the mucus barrier. METHODS: We used mice genetically deficient for a key enzyme for H(2)S production (cystathionine γ-lyase) and pharmacologically inhibited that enzyme during colitis in wild-type mice. We tested the effects of administering an H(2)S donor (diallyl disulfide) to rodents during hapten-induced colitis. Colonic microbiota biofilm was visualized by fluorescent in situ hybridization, and mucus granules were quantified with periodic acid-alcian blue staining. We exposed human microbiota biofilms and planktonic bacteria to H(2)S donors ex vivo to determine changes in their growth, viability, and biomass. RESULTS: Intestinal microbiota formed linear biofilms in the colon of healthy rodents. During colitis, microbiota biofilms were fragmented and mucus granule production decreased. Endogenous production of H(2)S had beneficial effects on establishment of microbiota biofilms and colonic mucus production. Therapeutic delivery of H(2)S into the colon reduced inflammation, restored the microbiota biofilm, and increased the production of mucus granules. In ex vivo human microbiota, H(2)S not only promoted biofilm formation but also reduced growth of planktonic bacteria. CONCLUSIONS: Our results suggest that H(2)S donors could be used therapeutically during colitis, facilitating correction of microbiota biofilm dysbiosis and mucus layer reconstitution.


Asunto(s)
Biopelículas/efectos de los fármacos , Colitis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Sulfuro de Hidrógeno/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Moco/metabolismo , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Sulfato de Dextran/toxicidad , Gasotransmisores/uso terapéutico , Humanos , Hibridación Fluorescente in Situ , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Moco/efectos de los fármacos , Ratas , Ratas Wistar
17.
FASEB J ; 29(4): 1591-602, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550470

RESUMEN

During a course of colitis, production of the gaseous mediator hydrogen sulfide (H2S) is markedly up-regulated at sites of mucosal damage and contributes significantly to healing and resolution of inflammation. The signaling mechanisms through which H2S promotes resolution of colitis are unknown. We hypothesized that the beneficial effects of H2S in experimental colitis are mediated via stabilization of hypoxia-inducible factor (HIF)-1α. The hapten dinitrobenzene sulfonic acid was used to induce colitis in rats and mice. This resulted in an elevated expression of the H2S-producing enzyme, cystathionine γ-lyase (CSE), and HIF-1α at sites of mucosal ulceration, and the expression of these 2 enzymes followed a similar pattern throughout the course of colitis. This represented a functionally important relationship because the loss of CSE-derived H2S production led to decreased HIF-1α stabilization and exacerbation of colitis. Furthermore, application of an H2S-releasing molecule, diallyl disulfide (DADS), stabilized colonic HIF-1α expression, up-regulated hypoxia-responsive genes, and reduced the severity of disease during peak inflammation. Importantly, the ability of DADS to promote the resolution of colitis was abolished when coadministered with an inhibitor of HIF-1α in vivo (PX-478). DADS was also able to maintain HIF-1α expression at a later point in colitis, when HIF-1α levels would have normally returned to control levels, and to enhance resolution. Finally, we found that HIF-1α stabilization inhibited colonic H2S production and may represent a negative feedback mechanism to prevent prolonged HIF-1α stabilization. Our findings demonstrate an important link between H2S and HIF-1α in the resolution of inflammation and injury during colitis and provide mechanistic insights into the therapeutic value of H2S donors.


Asunto(s)
Colitis/metabolismo , Sulfuro de Hidrógeno/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Compuestos Alílicos/farmacología , Animales , Bencenosulfonatos/toxicidad , Colitis/tratamiento farmacológico , Colitis/patología , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/metabolismo , Modelos Animales de Enfermedad , Disulfuros/farmacología , Expresión Génica , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Mostaza/farmacología , Fenilpropionatos/farmacología , Estabilidad Proteica , Ratas , Ratas Wistar , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
18.
PLoS One ; 9(10): e109087, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25289678

RESUMEN

Giardia duodenalis (syn. G. intestinalis, G. lamblia) is a predominant cause of waterborne diarrheal disease that may lead to post-infectious functional gastrointestinal disorders. Although Giardia-infected individuals could carry as much as 106 trophozoites per centimetre of gut, their intestinal mucosa is devoid of overt signs of inflammation. Recent studies have shown that in endemic countries where bacterial infectious diseases are common, Giardia infections can protect against the development of diarrheal disease and fever. Conversely, separate observations have indicated Giardia infections may enhance the severity of diarrheal disease from a co-infecting pathogen. Polymorphonuclear leukocytes or neutrophils (PMNs) are granulocytic, innate immune cells characteristic of acute intestinal inflammatory responses against bacterial pathogens that contribute to the development of diarrheal disease following recruitment into intestinal tissues. Giardia cathepsin B cysteine proteases have been shown to attenuate PMN chemotaxis towards IL-8/CXCL8, suggesting Giardia targets PMN accumulation. However, the ability of Giardia infections to attenuate PMN accumulation in vivo and how in turn this effect may alter the host inflammatory response in the intestine has yet to be demonstrated. Herein, we report that Giardia infection attenuates granulocyte tissue infiltration induced by intra-rectal instillation of Clostridium difficile toxin A and B in an isolate-dependent manner. This attenuation of granulocyte infiltration into colonic tissues paralled decreased expression of several cytokines associated with the recruitment of PMNs. Giardia trophozoite isolates that attenuated granulocyte infiltration in vivo also decreased protein expression of cytokines released from inflamed mucosal biopsy tissues collected from patients with active Crohn's disease, including several cytokines associated with PMN recruitment. These results demonstrate for the first time that certain Giardia infections may attenuate PMN accumulation by decreasing the expression of the mediators responsible for their recruitment.


Asunto(s)
Toxinas Bacterianas/efectos adversos , Colitis/etiología , Colitis/patología , Giardia/inmunología , Giardiasis/inmunología , Granulocitos/inmunología , Animales , Biopsia , Citocinas/metabolismo , Modelos Animales de Enfermedad , Enterotoxinas/efectos adversos , Giardiasis/parasitología , Granulocitos/patología , Humanos , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Mucosa Intestinal/patología , Masculino , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo
19.
Appl Environ Microbiol ; 79(24): 7745-54, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24096422

RESUMEN

The intestinal microbiota is a key determinant of gut homeostasis, which is achieved, in part, through regulation of antimicrobial peptide secretion. The aim of this study was to determine the efficiency by which members of the intestinal microbiota induce the antimicrobial peptide REGIII and to elucidate the underlying pathways. We showed that germfree mice have low levels of REGIII-γ in their ileum and colon compared to mice with different intestinal microbiota backgrounds. Colonization with a microbiota of low diversity (altered Schaedler flora) did not induce the expression of REGIII-γ as effectively as a complex community (specific pathogen free). Monocolonization with the probiotic Bifidobacterium breve, but not with the nonprobiotic commensal Escherichia coli JM83, upregulated REGIII-γ expression. Induction of REGIII-γ by B. breve was abrogated in mice lacking MyD88 and Ticam1 signaling. Both live and heat-inactivated B. breve but not spent culture medium from B. breve induced the expression of REGIII-α, the human ortholog and homolog of REGIII-γ, in human colonic epithelial cells (Caco-2). Taken together, the results suggest that REGIII-γ expression in the intestine correlates with the richness of microbiota composition. Also, specific bacteria such as Bifidobacterium breve NCC2950 effectively induce REGIII production in the intestine via the MyD88-Ticam1 pathway. Treatment with this probiotic may enhance the mucosal barrier and protect the host from infection and inflammation.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Biomarcadores de Tumor/metabolismo , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Expresión Génica , Lectinas Tipo C/metabolismo , Proteínas/metabolismo , Animales , Antígenos de Neoplasias/genética , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Biomarcadores de Tumor/genética , Células CACO-2 , Humanos , Lectinas Tipo C/genética , Ratones , Proteínas Asociadas a Pancreatitis , Proteínas/genética
20.
Am J Pathol ; 180(1): 141-52, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22067907

RESUMEN

Proteases and proteinase-activated receptor (PAR) activation are involved in several intestinal inflammatory conditions. We hypothesized that serine proteases and PAR activation could also modulate the intestinal injury induced by ischemia-reperfusion (I-R). C57Bl/6 mice were subjected to 90 minutes of intestinal ischemia followed or not by reperfusion. Sham-operated animals served as controls. After ischemia, plasma and tissue serine protease activity levels were increased compared to the activity measured in plasma and tissues from sham-operated mice. This increase was maintained or further enhanced after 2 and 5 hours of reperfusion, respectively. Trypsin (25 kDa) was detected in tissues both after ischemia and 2 hours of reperfusion. Treatment with FUT-175 (10 mg/kg), a potent serine protease inhibitor, increased survival after I-R, inhibited tissue protease activity, and significantly decreased intestinal myeloperoxidase (MPO) activity and chemokine and adhesion molecule expression. We investigated whether serine proteases modulate granulocyte recruitment by a PAR-dependent mechanism. MPO levels and adhesion molecule expression were significantly reduced in I-R groups pre-treated with the PAR(1) antagonist SCH-79797 (5 mg/kg) and in Par(2)(-/-)mice, compared, respectively, to vehicle-treated group and wild-type littermates. Thus, increased proteolytic activity and PAR activation play a pathogenic role in intestinal I-R injury. Inhibition of PAR-activating serine proteases could be beneficial to reduce post-ischemic intestinal inflammation.


Asunto(s)
Granulocitos/fisiología , Intestino Delgado/irrigación sanguínea , Isquemia/enzimología , Daño por Reperfusión/prevención & control , Inhibidores de Serina Proteinasa/farmacología , Animales , Benzamidinas , Quimiocinas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Granulocitos/enzimología , Guanidinas/farmacología , Isquemia/patología , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Inhibidores de Proteasas/farmacología , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Tripsina/metabolismo , alfa-Macroglobulinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA