Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Minerva Gastroenterol (Torino) ; 69(4): 523-528, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36943203

RESUMEN

BACKGROUND: Although proton pump inhibitor (PPI) drugs have considered able to induce small intestinal bacterial overgrowth (SIBO), no data are so far available from primary care (PC). We assessed the prevalence of SIBO and methane (CH4) production consequent to chronic PPI therapy using Lactulose Breath Test (LBT). Secondary aim was to explore the possible role of rifaximin in treating PPI-induced SIBO patients. METHODS: One hundred twenty-five gastroesophageal reflux disease patients, constantly treated with PPI for at least 6 months and undergoing to LBT, were retrospectively assessed. An age-matched control population (control) of 100 patients, which had not used PPI in the last 6 months, was also enrolled. In the PPI group, SIBO positive patients and CH4 producers were treated with rifaximin 1200 mg/daily for 14 days and re-checked with LBT one month after the end of treatment. The area under the curve (AUC) before and after treatment was also calculated for both SIBO positive patients and CH4 producers. RESULTS: In the PPI group, SIBO prevalence was significantly higher vs. controls (38/125 [30.4%] vs. 27/100 [27%], P<0.05). 77/125 (61.6%) PPI patients were found to be CH4 producers vs. 21/100 (21%) controls (P<0.05). Among SIBO patients in the PPI group, 34 (89.4%) were also CH4 producers vs. 17/27 (63%) controls (P<0.05). After treatment, LBT resulted negative in 15/22 SIBO patients (68.1%) (P<0.05) and in 18/34 CH4 producers (52.9%) (P<0.05). At the AUC analysis, an overall reduction of 54.2% for H2 in SIBO patients and of 47.7% for CH4 was assessed after rifaximin treatment (P<0.05). CONCLUSIONS: Our data showed that chronic use of PPI could be able to increase the prevalence of SIBO and to shift the intestinal microbial composition towards a CH4-producing flora. rifaximin could represent a useful therapeutical option for PPI-induced SIBO and for modulating CH4-producing flora.


Asunto(s)
Intestino Delgado , Inhibidores de la Bomba de Protones , Humanos , Niño , Rifaximina/uso terapéutico , Inhibidores de la Bomba de Protones/efectos adversos , Estudios Retrospectivos , Prevalencia , Lactulosa/uso terapéutico , Metano , Atención Primaria de Salud
2.
Artículo en Inglés | MEDLINE | ID: mdl-34278753

RESUMEN

In inflammatory bowel disease (IBD), the loss of immune tolerance against gut microbiota causes chronic inflammation and the progressive accumulation of organ damage in genetically susceptible individuals. In the elderly, IBD is often characterized by a different disease behaviour when compared with paediatric and young adult disease. Besides disease behaviour, another aspect of the multifaceted impact of age on elderly IBD course is increased susceptibility to infections. In this context, age-of-onset-dependent IBD behaviour and clinical course are two major contributors to immune system senescence and change of gut microbiota in older subjects. Here, we review the available literature linking immunosenescence and age-dependent changes in the gut microbiota composition to IBD pathogenesis speculating on their possible implications in disease expression in this age class.

3.
Expert Opin Biol Ther ; 20(4): 343-351, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32083498

RESUMEN

Introduction: Established evidence suggests that gut microbiota plays a role in ulcerative colitis (UC). Fecal microbiota transplantation (FMT) is clearly recognized as a highly effective treatment for patients with recurrent Clostridium difficile infection and has been investigated also in patients with UC, with promising results.Areas covered: Literature review was performed to select publications concerning current evidence on the role of gut microbiota in the pathogenesis of UC, and on the effectiveness of FMT in this disorder.Expert opinion: The randomized controlled trials published investigating the use of FMT suggested a potential role for FMT in the treatment of mild to moderate UC. However, given several unanswered questions regarding donor selection, dose, route of administration and duration of therapy, this is not yet recommended as a viable therapy option. FMT has allowed for more in depth investigation with regards to the role the gut microbiota may be playing in UC. This knowledge is critical to identifying where FMT may appropriately fit in the UC treatment paradigm. As our understanding of the role the microbiome plays in this chronic disease, FMT, and then eventually defined microbes, will hopefully serve in a complementary role to conventional IBD therapies.


Asunto(s)
Colitis Ulcerosa/terapia , Trasplante de Microbiota Fecal , Colitis Ulcerosa/patología , Heces/microbiología , Microbioma Gastrointestinal , Humanos , Interleucina-1/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Liver Int ; 40(4): 878-888, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31951082

RESUMEN

BACKGROUND & AIMS: Alcohol use disorder (AUD) represents the most common cause of liver disease. The gut microbiota plays a critical role in the progression of alcohol-related liver damage. Aim of this study was to characterize the gut microbial composition and function in AUD patients with alcohol-associated liver disease (AALD). METHODS: This study included 36 AUD patients (14 with cirrhosis) who were active drinkers and an equal number of matched controls. Stool microbial composition, serum levels of lipopolysaccharide, cytokines/chemokines and gut microbiota functional profile were assessed. RESULTS: AUD patients had a decreased microbial alpha diversity as compared to controls (0.092 vs 0.130, P = .047) and a specific gut microbial signature. The reduction of Akkermansia and the increase in Bacteroides were able to identify AUD patients with an accuracy of 93.4%. Serum levels of lipopolysaccharide (4.91 vs 2.43, P = .009) and pro-inflammatory mediators (tumour necrosis factor alpha 60.85 vs 15.08, P = .001; interleukin [IL] 1beta 4.43 vs 1.72, P = .0001; monocyte chemoattractant protein 1 225.22 vs 16.43, P = .006; IL6 1.87 vs 1.23, P = .008) were significantly increased in AUD patients compared to controls and in cirrhotic patients compared to non-cirrhotic ones (IL6 3.74 vs 1.39, P = .019; IL8 57.60 vs 6.53, P = .004). The AUD-associated gut microbiota showed an increased expression of gamma-aminobutyric acid (GABA) metabolic pathways and energy metabolism. CONCLUSIONS: AUD patients present a specific gut microbial fingerprint, associated with increased endotoxaemia, systemic inflammatory status and functional alterations that may be involved in the progression of the AALD and in the pathogenesis of AUD.


Asunto(s)
Alcoholismo , Microbioma Gastrointestinal , Hepatopatías Alcohólicas , Alcoholismo/complicaciones , Heces , Humanos , Cirrosis Hepática
5.
World J Gastroenterol ; 25(12): 1465-1477, 2019 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-30948910

RESUMEN

BACKGROUND: Anti-tumor necrosis factor α (TNFα) represents the best therapeutic option to induce mucosal healing and clinical remission in patients with moderate-severe ulcerative colitis. On the other side gut microbiota plays a crucial role in pathogenesis of ulcerative colitis but few information exists on how microbiota changes following anti-TNFα therapy and on microbiota role in mucosal healing. AIM: To elucidate whether gut microbiota and immune system changes appear following anti TNFα therapy during dextran sulfate sodium (DSS) colitis. METHODS: Eighty C57BL/6 mice were divided into four groups: "No DSS", "No DSS + anti-TNFα", "DSS" and "DSS + anti-TNFα". "DSS" and "DSS + anti-TNFα" were treated for 5 d with 3% DSS. At day 3, mice whithin "No DSS+anti-TNFα" and "DSS+anti-TNFα" group received 5 mg/kg of an anti-TNFα agent. Forty mice were sacrificed at day 5, forty at day 12, after one week of recovery post DSS. The severity of colitis was assessed by a clinical score (Disease Activity Index), colon length and histology. Bacteria such as Bacteroides, Clostridiaceae, Enterococcaceae and Fecalibacterium prausnitzii (F. prausnitzii) were evaluated by quantitative PCR. Type 1 helper T lymphocytes (Th1), type 17 helper T lymphocytes (Th17) and CD4+ regulatory T lymphocytes (Treg) distributions in the mesenteric lymph node (MLN) were studied by flow cytometry. RESULTS: Bacteria associated with a healthy state (i.e., such as Bacteroides, Clostridiaceae and F. prausnitzii) decreased during colitis and increased in course of anti-TNFα treatment. Conversely, microorganisms belonging to Enterococcaceae genera, which are linked to inflammatory processes, showed an opposite trend. Furthermore, in colitic mice treated with anti-TNFα microbial changes were associated with an initial increase (day 5 of the colitis) in Treg cells and a consequent decrease (day 12 post DSS) in Th1 and Th17 frequency cells. Healthy mice treated with anti-TNFα showed the same histological, microbial and immune features of untreated colitic mice. "No DSS + anti-TNFα" group showed a lymphomononuclear infiltrate both at 5th and 12th d at hematoxylin and eosin staining, an increase of in Th1 and Th17 frequency at day 12, an increase of Enterococcaceae at day 5, a decrease of Bacteroides and Clostridiaceae at day 12. CONCLUSION: Anti-TNFα treatment in experimental model of colitis improves disease activity but it is associated to an increase in Th17 pathway together with gut microbiota alteration.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Fármacos Gastrointestinales/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Células Th17/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Bacterias/efectos de los fármacos , Bacterias/inmunología , Bacterias/aislamiento & purificación , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/inmunología , Colon/efectos de los fármacos , Colon/inmunología , Colon/microbiología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Humanos , Infliximab/efectos adversos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología
6.
Proc Natl Acad Sci U S A ; 115(40): E9362-E9370, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30224451

RESUMEN

Defective and/or delayed wound healing has been implicated in the pathogenesis of several chronic inflammatory disorders, including inflammatory bowel disease (IBD). The resolution of inflammation is particularly important in mucosal organs, such as the gut, where restoration of epithelial barrier function is critical to reestablish homeostasis with the interfacing microenvironment. Although IL-33 and its receptor ST2/ILRL1 are known to be increased and associated with IBD, studies using animal models of colitis to address the mechanism have yielded ambiguous results, suggesting both pathogenic and protective functions. Unlike those previously published studies, we focused on the functional role of IL-33/ST2 during an extended (2-wk) recovery period after initial challenge in dextran sodium sulfate (DSS)-induced colitic mice. Our results show that during acute, resolving colitis the normal function of endogenous IL-33 is protection, and the lack of either IL-33 or ST2 impedes the overall recovery process, while exogenous IL-33 administration during recovery dramatically accelerates epithelial restitution and repair, with concomitant improvement of colonic inflammation. Mechanistically, we show that IL-33 stimulates the expression of a network of microRNAs (miRs) in the Caco2 colonic intestinal epithelial cell (IEC) line, especially miR-320, which is increased by >16-fold in IECs isolated from IL-33-treated vs. vehicle-treated DSS colitic mice. Finally, IL-33-dependent in vitro proliferation and wound closure of Caco-2 IECs is significantly abrogated after specific inhibition of miR-320A. Together, our data indicate that during acute, resolving colitis, IL-33/ST2 plays a crucial role in gut mucosal healing by inducing epithelial-derived miR-320 that promotes epithelial repair/restitution and the resolution of inflammation.


Asunto(s)
Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-33/metabolismo , Mucosa Intestinal/fisiología , MicroARNs/metabolismo , Regeneración , Enfermedad Aguda , Animales , Células CACO-2 , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Sulfato de Dextran/toxicidad , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Ratones , Ratones Noqueados , MicroARNs/genética
7.
Expert Rev Gastroenterol Hepatol ; 12(9): 899-906, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30040500

RESUMEN

INTRODUCTION: Helicobacter pylori (Hp) infection produces drastic changes in the gastric microenvironment, which, in turn, influence the gastric microbiota composition and might be correlated with large intestinal microbiota changes. This excellent perturbing actor could trigger important modifications in the homeostatic functions exerted by gut commensals leading to a new gastrointestinal balance. At the same time, the therapeutic strategies used to eradicate Hp can modulate this physiological symbiosis, but can be also conversely affected by its properties. Area covered: The purpose of this review is to explore the reciprocal interplay between Hp infection and gut microbiota and analyze how microbial changes can influence the management of Hp eradication therapies. Expert commentary: While many studies have described Hp-dependent gut microbiota alterations, their clinical implications are only partially clear, as well as the mechanism of actions that sustain these processes. This represents a clear challenge for future research projects that will also need to understand which role is exerted by viruses, parasites, and yeasts.


Asunto(s)
Antibacterianos/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Intestinos/efectos de los fármacos , Inhibidores de la Bomba de Protones/uso terapéutico , Estómago/efectos de los fármacos , Antibacterianos/efectos adversos , Quimioterapia Combinada , Disbiosis , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Inhibidores de la Bomba de Protones/efectos adversos , Estómago/microbiología , Simbiosis , Resultado del Tratamiento
8.
Expert Opin Investig Drugs ; 27(6): 543-551, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29865875

RESUMEN

INTRODUCTION: The gastrointestinal tract acts as a functional unit organized as a semipermeable multilayer system, in which commensal gut microbiota represents the anatomical barrier. Recently, several studies have highlighted the involvement of gut microbiota in inflammatory bowel diseases (IBD) pathogenesis, in sustaining gut barrier chronic inflammation, and in conditioning disease course and therapeutical response. This evidence provides a rationale for treating patients with gut microbiota modifiers. Among these, Rifaximin represents a non-traditional antibiotic able to act as a 'eubiotic' on intestinal barrier. AREAS COVERED: The purpose of this narrative review is to explore the impact of Rifaximin on gut barrier and gut microbiota in IBD, in particular in Crohn's disease (CD), and to analyze its potential therapeutic applications. EXPERT OPINION: The possibility of a beneficial activity of Rifaximin in chronic intestinal inflammation and CD has been debated and evaluated with different studies having obtained promising but still preliminary data. Larger trials are therefore needed. This gut-specific antibiotic could represent an alternative to systemic antibiotics thanks to its favorable safety profile and promising efficacy data. Rifaximin could exert, when appropriate, a synergic effect with immunomodulators in IBD, acting on both the microbial and the immunological sides of gut barrier impairment.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Fármacos Gastrointestinales/administración & dosificación , Rifamicinas/administración & dosificación , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/efectos adversos , Antiinfecciosos/farmacología , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/fisiopatología , Sinergismo Farmacológico , Fármacos Gastrointestinales/efectos adversos , Fármacos Gastrointestinales/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Factores Inmunológicos/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/fisiopatología , Rifamicinas/efectos adversos , Rifamicinas/farmacología , Rifaximina
9.
Inflamm Bowel Dis ; 24(10): 2198-2210, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29788266

RESUMEN

Background: Magnesium (Mg) is essential for human health and is absorbed mainly in the intestine. In view of the likely occurrence of an Mg deficit in inflammatory bowel disease (IBD) and the documented role of Mg in modulating inflammation, the present study addresses whether Mg availability can affect the onset and progression of intestinal inflammation. Methods: To study the correlation between Mg status and disease activity, we measured magnesemia by atomic absorption spectroscopy in a cohort of IBD patients. The effects of dietary Mg modulation were assessed in a murine model of dextran sodium sulfate (DSS)-induced colitis by monitoring magnesemia, weight, fecal occult blood, diarrhea, colon length, and histology. Expression of the transient receptor potential melastatin (TRPM) 6 channel was assessed by real-time reverse transcription polymerase chain reaction and immunohistochemistry in murine colon tissues. The effect of Mg on epithelial barrier formation/repair was evaluated in human colon cell lines. Results: Inflammatory bowel disease patients presented with a substantial Mg deficit, and serum Mg levels were inversely correlated with disease activity. In mice, an Mg-deficient diet caused hypomagnesemia and aggravated DSS-induced colitis. Colitis severely compromised intestinal Mg2+ absorption due to mucosal damage and reduction in TRPM6 expression, but Mg supplementation resulted in better restoration of mucosal integrity and channel expression. Conclusions: Our results highlight the importance of evaluating and correcting magnesemia in IBD patients. The murine model suggests that Mg supplementation may represent a safe and cost-effective strategy to reduce inflammation and restore normal mucosal function.


Asunto(s)
Colitis Ulcerosa/complicaciones , Colitis/prevención & control , Enfermedad de Crohn/complicaciones , Dieta , Hipocalcemia/metabolismo , Deficiencia de Magnesio/congénito , Magnesio/administración & dosificación , Canales Catiónicos TRPM/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/fisiopatología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/fisiopatología , Sulfato de Dextran/toxicidad , Femenino , Estudios de Seguimiento , Humanos , Hipocalcemia/etiología , Hipocalcemia/patología , Magnesio/metabolismo , Deficiencia de Magnesio/etiología , Deficiencia de Magnesio/metabolismo , Deficiencia de Magnesio/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pronóstico , Canales Catiónicos TRPM/genética , Adulto Joven
10.
Front Microbiol ; 9: 675, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29686658

RESUMEN

Pediatric acute-onset neuropsychiatric syndrome (PANS) and pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections syndrome (PANDAS) are conditions that impair brain normal neurologic function, resulting in the sudden onset of tics, obsessive-compulsive disorder, and other behavioral symptoms. Recent studies have emphasized the crosstalk between gut and brain, highlighting how gut composition can influence behavior and brain functions. Thus, the present study investigates the relationship between PANS/PANDAS and gut microbiota ecology. The gut composition of a cohort of 30 patients with PANS/PANDAS was analyzed and compared to control subjects using 16S rRNA-based metagenomics. Data were analyzed for their α- and ß-diversity; differences in bacterial distribution were detected by Wilcoxon and LEfSe tests, while metabolic profile was predicted via PICRUSt software. These analyses demonstrate the presence of an altered bacterial community structure in PANS/PANDAS patients with respect to controls. In particular, ecological analysis revealed the presence of two main clusters of subjects based on age range. Thus, to avoid age bias, data from patients and controls were split into two groups: 4-8 years old and >9 years old. The younger PANS/PANDAS group was characterized by a strong increase in Bacteroidetes; in particular, Bacteroides, Odoribacter, and Oscillospira were identified as potential microbial biomarkers of this composition type. Moreover, this group exhibited an increase of several pathways concerning the modulation of the antibody response to inflammation within the gut as well as a decrease in pathways involved in brain function (i.e., SCFA, D-alanine and tyrosine metabolism, and the dopamine pathway). The older group of patients displayed a less uniform bacterial profile, thus impairing the identification of distinct biomarkers. Finally, Pearson's analysis between bacteria and anti-streptolysin O titer reveled a negative correlation between genera belonging to Firmicutes phylum and anti-streptolysin O while a positive correlation was observed with Odoribacter. In conclusion, this study suggests that streptococcal infections alter gut bacterial communities leading to a pro-inflammatory status through the selection of specific bacterial strains associated with gut inflammation and immune response activation. These findings highlight the possibility of studying bacterial biomarkers associated with this disorder and might led to novel potential therapeutic strategies.

11.
Minerva Gastroenterol Dietol ; 63(4): 373-384, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28293937

RESUMEN

Major advances have occurred in the knowledge of the pathogenesis of inflammatory bowel disease (IBD) over the last decade, and perhaps the most major, and clinically advantageous of these advances has been the discovery of the microbiome as a key multifaceted component of inflammation. The gut microbiome is the largest known group of cells in the body, and is now recognized as an organ in its own right. Initial studies looking at a possible role of bacterial manipulation of the immune system in IBD, looked at identifying a specific bacterial species, and were not representative of a feasible model of inflammation in IBD overall. More recently there has been a shift towards the concept of dysbiosis, and the acceptance that a number of bacterial factors interact with the immune system in order for inflammation to occur. In the present review we will focus on past perspective of the role of microbiota in IBD, current evidences about dysbiosis in IBD and also the main therapeutic modalities to affect IBD by affecting gut microbiota: probiotics, prebiotics, fecal microbiota transplantation and emerging dietary intervention.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Disbiosis/terapia , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Resultado del Tratamiento
12.
PLoS One ; 12(3): e0174121, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28301579

RESUMEN

Patients with inflammatory bowel disease (IBD) are at increased risk for developing colorectal cancer. Evidence suggests that colonic dysplasia and colitis-associated cancer (CAC) are often linked to repeated cycles of epithelial cell injury and repair in the context of chronic production of inflammatory cytokines. Several mouse models of CAC have been proposed, including chemical induction through exposure to dextran sulfate sodium (DSS) with the genotoxic agents azoxymethane (AOM), 1,2-dymethylhydrazine (DHM) or targeted genetic mutations. However, such models are usually performed on healthy animals that usually lack the underlying genetic predisposition, immunological dysfunction and dysbiosis characteristic of IBD. We have previously shown that inbred SAMP1/YitFc (SAMP) mice develop a progressive Crohn's disease (CD)-like ileitis in the absence of spontaneous colitis. We hypothesize that SAMP mice may be more susceptible to colonic tumorigenesis due to their predisposition to IBD. To test this hypothesis, we administered AOM/DSS to IBD-prone SAMP and their non-inflamed parental control strain, AKR mice. Our results showed that AOM/DSS treatment enhanced the susceptibility of colitis in SAMP compared to AKR mice, as assessed by endoscopic and histologic inflammatory scores, daily weight loss and disease activity index (DAI), during and after DSS administration. SAMP mice also showed increased colonic tumorigenesis, resulting in the occurrence of intramucosal carcinoma and a higher incidence of high-grade dysplasia and tumor burden. These phenomena occurred even in the absence of AOM and only upon repeated cycles of DSS. Taken together, our data demonstrate a heightened susceptibility to colonic inflammation and tumorigenesis in AOM/DSS-treated SAMP mice with CD-like ileitis. This novel model represents a useful tool to investigate relevant mechanisms of CAC, as well as for pre-clinical testing of potential IBD and colon cancer therapeutics.


Asunto(s)
Neoplasias del Colon/etiología , Enfermedad de Crohn/complicaciones , Modelos Animales de Enfermedad , Ileítis/complicaciones , Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Animales , Neoplasias del Colon/genética , Enfermedad de Crohn/genética , Ileítis/genética , Ratones
13.
Inflamm Bowel Dis ; 23(3): 392-403, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28146004

RESUMEN

BACKGROUND: A large body of evidence supports a central role of TLR5 and its natural ligand, flagellin, in Crohn's disease (CD), with the precise mechanism(s) still unresolved. METHODS: We investigated the role of flagellin/TLR5 in SAMP1/YitFc (SAMP) mice, a spontaneous model of Crohn's disease-like ileitis. RESULTS: Ileal Tlr5 and serum antiflagellin IgG antibodies were increased in SAMP before the onset of inflammation and during established disease; these trends were abrogated in the absence of colonizing commensal bacteria. Irradiated SAMP receiving either wild-type (AKR) or SAMP bone marrow (BM) developed severe ileitis and displayed increased ileal Tlr5 compared with AKR recipients of either SAMP or AKR bone marrow, neither of which conferred ileitis, suggesting that elevated TLR5 in native SAMP is derived primarily from a nonhematopoietic (e.g., epithelial) source. Indeed, ileal epithelial TLR5 in preinflamed SAMP was increased compared with age-matched AKR and germ-free SAMP. TLR5-specific ex vivo activation of SAMP ileal tissues decreased epithelial barrier resistance, indicative of increased permeability, and was accompanied by altered expression of the tight junction proteins, claudin-3, occludin, and zonula occludens-1. CONCLUSIONS: Our results provide evidence that aberrant, elevated TLR5 expression is present in the ileal epithelium of SAMP mice, is augmented in the presence of the gut microbiome, and that TLR5 activation in response to bacterial flagellin results in a deficiency to maintain appropriate epithelial barrier integrity. Together, these findings represent a potential mechanistic pathway leading to the exacerbation and perpetuation of chronic gut inflammation in experimental ileitis and possibly, in patients with Crohn's disease.


Asunto(s)
Ileítis/inmunología , Mucosa Intestinal/metabolismo , Receptor Toll-Like 5/fisiología , Animales , Enfermedad de Crohn/inmunología , Modelos Animales de Enfermedad , Flagelina/inmunología , Flagelina/metabolismo , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos AKR , Permeabilidad , Proteínas de Uniones Estrechas/metabolismo , Receptor Toll-Like 5/inmunología
14.
World J Gastroenterol ; 22(44): 9727-9733, 2016 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-27956796

RESUMEN

AIM: To explore the influence of Infliximab (IFX) on cancer progression in a murine model of colonic cancer associated to chronic colitis. METHODS: AOM/DSS model was induced in C57BL/6 mice. Mice were injected with IFX (5 mg/kg) during each DSS cycle while control mice received saline. Body weight, occult blood test and stool consistency were measured to calculate the disease activity index (DAI). Mice were sacrificed at week 10 and colons were analyzed macroscopically and microscopically for number of cancers and degree of inflammation. MTT assay was performed on CT26 to evaluate the potential IFX role on metabolic activity and proliferation. Cells were incubated with TNF-α or IFX or TNF-α plus IFX, and cell vitality was evaluated after 6, 24 and 48 h. The same setting was used after pre-incubation with TNF-α for 24 h. RESULTS: IFX significantly reduced DAI and body weight loss in mice compared with controls, preserving also colon length at sacrifice. Histological score was also reduced in treated mice. At macroscopic analysis, IFX treated mice showed a lower number of tumor lesions compared to controls. This was confirmed at microscopic analysis, although differences were not statistically significant. In vitro, IFX treated CT26 maintained similar proliferation ability at MTT test, both when exposed to IFX alone and when associated to TNF-α. CONCLUSION: IFX did not increase colonic cancer risk in AOM-DSS model of cancer on chronic colitis nor influence directly the proliferation of murine colon cancer epithelial cells.


Asunto(s)
Antiinflamatorios/farmacología , Colitis/prevención & control , Colon/efectos de los fármacos , Neoplasias del Colon/etiología , Fármacos Gastrointestinales/farmacología , Infliximab/farmacología , Animales , Antiinflamatorios/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/patología , Colon/patología , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Sulfato de Dextran , Modelos Animales de Enfermedad , Fármacos Gastrointestinales/toxicidad , Infliximab/toxicidad , Ratones Endogámicos C57BL , Factores de Tiempo , Factor de Necrosis Tumoral alfa/farmacología
15.
Expert Rev Gastroenterol Hepatol ; 10(8): 943-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27291780

RESUMEN

INTRODUCTION: The intestinal barrier is a complex system responsible for the host health. Many gastrointestinal and extra-intestinal diseases are associated to gut barrier disruption. An increasing interest on nutritional supplements and functional foods focused on the hypothesis that specific prebiotics and probiotics may modulate and interact with gut barrier, re-establishing gut homeostasis. AREAS COVERED: The application of preparations containing B. clausii in the treatment or prevention of gut phisiology impairment has been largely supported in the last years and has driven its clinical applications. This review focuses on B. clausii clinical applications and speculates on the possible interactions among B. clausii, gut barrier and immune system and on the consequences of this interplay in modulating human health. Expert commentary: Its favorable effects have been linked to several properties, such as antimicrobial and immunomodulatory activity, regulation of cell growth and differentiation, cell-cell signaling, cell adhesion, signal transcription and transduction, production of vitamins and gut protection from genotoxic agents. In this scenario, future studies will need to better clarify its mechanisms of action and focus on the possible role of B. clausii in modulating gut immune system.


Asunto(s)
Bacillus clausii/fisiología , Enfermedades Gastrointestinales/terapia , Microbioma Gastrointestinal , Intestinos/microbiología , Animales , Bacillus clausii/inmunología , Disbiosis , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/fisiopatología , Homeostasis , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/fisiopatología , Probióticos
16.
Inflamm Bowel Dis ; 22(7): 1708-12, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27206017

RESUMEN

Gut virome has been shown to yield some beneficial effects on humans, being deeply involved in physiology, inflammation, immunity, and disease. Together with transkingdom interactions, it can interplay with genetic variation in the host to establish specific phenotypes. These interactions can lead to phenotypes not observed with either the virus or the host variation alone. Unfavorable alteration of gut virome composition has been implicated in chronic, and perhaps also systemic, immune disorders, such as in the pathogenesis of inflammatory bowel disease. This review focuses on what is currently known regarding the role of commensal gut virome in chronic gut inflammation, and speculate on the important translational implications in regard to gut virome modulation in inflammatory bowel disease with the end goal of promoting gut health.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal/virología , Enfermedades Inflamatorias del Intestino/virología , Tracto Gastrointestinal/fisiopatología , Humanos , Enfermedades Inflamatorias del Intestino/fisiopatología
17.
Dig Dis ; 34(3): 269-78, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27027301

RESUMEN

Antibiotics are mainly used in clinical practice for their activity against pathogens, but they also alter the composition of commensal gut microbial community. Rifaximin is a non-absorbable antibiotic with additional effects on the gut microbiota about which very little is known. It is still not clear to what extent rifaximin can be able to modulate gut microbiota composition and diversity in different clinical settings. Studies based on culture-dependent techniques revealed that rifaximin treatment promotes the growth of beneficial bacteria, such as Bifidobacteria and Lactobacilli. Accordingly, our metagenomic analysis carried out on patients with different gastrointestinal and liver diseases highlighted a significant increase in Lactobacilli after rifaximin treatment, persisting in the short time period. This result was independent of the disease background and was not accompanied by a significant alteration of the overall gut microbial ecology. This suggests that rifaximin can exert important eubiotic effects independently of the original disease, producing a favorable gut microbiota perturbation without changing its overall composition and diversity.


Asunto(s)
Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Rifamicinas/farmacología , Antibacterianos/uso terapéutico , ADN Bacteriano/aislamiento & purificación , Humanos , Lactobacillus/efectos de los fármacos , Rifamicinas/administración & dosificación , Rifaximina
18.
Mini Rev Med Chem ; 16(3): 179-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26643042

RESUMEN

The gastrointestinal tract is a complex and dynamic network where an intricate and mutualistic symbiosis modulates the relationship between the host and the microbiota in order to establish and ensure gut homeostasis. Every day, thousands of compounds derived from food and microorganisms come in contact with the intestinal mucosa. This interaction requires a complex defense system that separates intestinal contents from the host tissues, regulates nutrient absorption, and allows tolerance between the resident bacterial flora and the mucosal immune system, while inhibiting translocation of infectious agents to the inner tissues. Unfavorable alteration of microbiota composition has been implicated in hepatic, gastrointestinal, and perhaps also systemic disorders. In this scenario, gut microbiota modulation represents an intriguing field and can be obtained by several approaches, including antibiotics, pro- and pre-biotics supplementation. Among antibiotics, Rifaximin seems to be a promising antibiotic to treat conditions related to gut microbiota imbalance and to potentially modulate intestinal homeostasis. This review focuses on what is currently known regarding the possible role of Rifaximin in restoring normal gut immune physiology and a healthy gut-liver axis. Detailed mechanistic studies will improve the development of targeted therapies that may shape gut microflora composition with the end goal of promoting gut health.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Rifamicinas/farmacología , Homeostasis/efectos de los fármacos , Humanos , Modelos Biológicos , Rifaximina
19.
Best Pract Res Clin Gastroenterol ; 28(6): 995-1002, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25439066

RESUMEN

The gastrointestinal (GI) tract is a complex and dynamic network with interplay between various gut mucosal cells and their defence molecules, the immune system, food particles, and the resident microbiota. This ecosystem acts as a functional unit organized as a semipermeable multi-layer system that allows the absorption of nutrients and macromolecules required for human metabolic processes and, on the other hand, protects the individual from potentially invasive microorganisms. Commensal microbiota and the host are a unique entity in a continuum along the GI tract, every change in one of these players is able to modify the whole homeostasis. In the stomach, Helicobacter pylori is a gram-negative pathogen that is widespread all over the world, infecting more than 50% of the world's population. In this scenario, H. pylori infection is associated with changes in the gastric microenvironment, which in turn affects the gastric microbiota composition, but also might trigger large intestinal microbiota changes. It is able to influence all the vital pathways of human system and also to influence microbiota composition along the GI tract. This can cause a change in the normal functions exerted by intestinal commensal microorganisms leading to a new gastrointestinal physiological balance. This review focuses and speculates on the possible interactions between gastric microorganisms and intestinal microbiota and on the consequences of this interplay in modulating gut health.


Asunto(s)
Tracto Gastrointestinal/microbiología , Helicobacter pylori/fisiología , Intestinos/microbiología , Microbiota/fisiología , Estómago/microbiología , Mucosa Gástrica/fisiología , Tracto Gastrointestinal/fisiopatología , Humanos
20.
Gut Pathog ; 5(1): 23, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23941657

RESUMEN

The gastrointestinal tract is a complex and dynamic network where an intricate and mutualistic symbiosis modulates the relationship between the host and the microbiota in order to establish and ensure gut homeostasis. Commensal Clostridia consist of gram-positive, rod-shaped bacteria in the phylum Firmicutes and make up a substantial part of the total bacteria in the gut microbiota. They start to colonize the intestine of breastfed infants during the first month of life and populate a specific region in the intestinal mucosa in close relationship with intestinal cells. This position allows them to participate as crucial factors in modulating physiologic, metabolic and immune processes in the gut during the entire lifespan, by interacting with the other resident microbe populations, but also by providing specific and essential functions. This review focus on what is currently known regarding the role of commensal Clostridia in the maintenance of overall gut function, as well as touch on their potential contribution in the unfavorable alteration of microbiota composition (dysbiosis) that has been implicated in several gastrointestinal disorders. Commensal Clostridia are strongly involved in the maintenance of overall gut function. This leads to important translational implications in regard to the prevention and treatment of dysbiosis, to drug efficacy and toxicity, and to the development of therapies that may modulate the composition of the microflora, capitalizing on the key role of commensal Clostridia, with the end goal of promoting gut health.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA