Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 513, 2023 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-37516837

RESUMEN

BACKGROUND: Chronic fatigue syndrome (CFS) is a disabling illness of unknown aetiology. Disruption of gut microbiota may play a role in several neurological disorders. In this study, the effect of faecal microbiota transplantation (FMT) on fatigue severity and health-related quality of life (HRQOL) in patients with CFS was evaluated. METHODS: Randomized, placebo-controlled pilot trial. Patients and researchers were blinded to treatment assignment. 11 patients with CFS (10 female and 1 male, mean age 42.2 years and mean duration of CFS 6.3 years) were randomly assigned to receive either FMT from a universal donor (n = 5) or autologous FMT (n = 6) via colonoscopy. Patients' HRQOL was assessed by using visual analog scale (VAS) and self-reporting questionnaires Modified Fatigue Impact Scale (MFIS), 15D and EQ-5D-3L. Patients' HRQOL was evaluated at baseline, and 1 and 6 months after the FMT. RESULTS: The baseline VAS scores in the FMT and placebo groups were 62.4 and 76.0 (p = 0.29). 1-month scores were 60.0 and 73.7 and 6-months scores 72.8 and 69.5, respectively. Total MFIS scores in the FMT and placebo groups were 59.6 and 61.0 at the baseline (p = 0.80), 53.5 and 62.0 at 1 month and 58.6 and 56.2 at 6 months. Compared to the baseline scores, differences at 1 and 6 months were statistically insignificant both in VAS and in MFIS. The 15D and EQ-5D-3L profiles did not change after the FMT or placebo. FMT-related adverse events were not reported. CONCLUSION: FMT was safe but did not relieve symptoms or improve the HRQOL of patients with CFS. Small number of study subjects limits the generalizability of these results. Trial Registration ClinicalTrials.gov Identifier NCT04158427, https://register. CLINICALTRIALS: gov , date of registration 08/08/2019.


Asunto(s)
Síndrome de Fatiga Crónica , Humanos , Femenino , Masculino , Adulto , Síndrome de Fatiga Crónica/terapia , Trasplante de Microbiota Fecal , Proyectos Piloto , Calidad de Vida , Método Doble Ciego
2.
Gut ; 69(9): 1555-1563, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32620549

RESUMEN

The COVID-19 pandemic has led to an exponential increase in SARS-CoV-2 infections and associated deaths, and represents a significant challenge to healthcare professionals and facilities. Individual countries have taken several prevention and containment actions to control the spread of infection, including measures to guarantee safety of both healthcare professionals and patients who are at increased risk of infection from COVID-19. Faecal microbiota transplantation (FMT) has a well-established role in the treatment of Clostridioides difficile infection. In the time of the pandemic, FMT centres and stool banks are required to adopt a workflow that continues to ensure reliable patient access to FMT while maintaining safety and quality of procedures. In this position paper, based on the best available evidence, worldwide FMT experts provide guidance on issues relating to the impact of COVID-19 on FMT, including patient selection, donor recruitment and selection, stool manufacturing, FMT procedures, patient follow-up and research activities.


Asunto(s)
Infecciones por Clostridium/terapia , Infecciones por Coronavirus , Selección de Donante , Trasplante de Microbiota Fecal/métodos , Gastroenterología , Pandemias , Selección de Paciente , Neumonía Viral , Betacoronavirus , COVID-19 , Gestión del Cambio , Infecciones por Clostridium/microbiología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Gastroenterología/organización & administración , Gastroenterología/tendencias , Microbioma Gastrointestinal , Humanos , Control de Infecciones/métodos , Control de Infecciones/normas , Pandemias/prevención & control , Neumonía Viral/epidemiología , Neumonía Viral/prevención & control , Ajuste de Riesgo/métodos , Ajuste de Riesgo/normas , SARS-CoV-2
3.
Int J Mol Sci ; 21(17)2020 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842596

RESUMEN

Inflammatory bowel diseases (IBD), ulcerative colitis (UC) and Crohn's disease (CD), are chronic debilitating disorders of unknown etiology. Over 200 genetic risk loci are associated with IBD, highlighting a key role for immunological and epithelial barrier functions. Environmental factors account for the growing incidence of IBD, and microbiota are considered as an important contributor. Microbiota dysbiosis can lead to a loss of tolerogenic immune effects and initiate or exacerbate inflammation. We aimed to study colonic mucosal microbiota and the expression of selected host genes in pediatric UC. We used high-throughput 16S rDNA sequencing to profile microbiota in colonic biopsies of pediatric UC patients (n = 26) and non-IBD controls (n = 27). The expression of 13 genes, including five for antimicrobial peptides, in parallel biopsies was assessed with qRT-PCR. The composition of microbiota between UC and non-IBD differed significantly (PCoA, p = 0.001). UC children had a decrease in Bacteroidetes and an increase in several family-level taxa including Peptostreptococcaceae and Enterobacteriaceae, which correlated negatively with the expression of antimicrobial peptides REG3G and DEFB1, respectively. Enterobacteriaceae correlated positively with the expression siderophore binding protein LCN2 and Betaproteobacteria negatively with DEFB4A expression. The results indicate that reciprocal interaction of epithelial microbiota and defense mechanisms play a role in UC.


Asunto(s)
Colitis Ulcerosa/microbiología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/fisiología , Proteínas Citotóxicas Formadoras de Poros/genética , Adolescente , Bacteroidetes/genética , Estudios de Casos y Controles , Niño , Preescolar , ADN Ribosómico , Enterobacteriaceae/genética , Finlandia , Microbioma Gastrointestinal/genética , Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/patología , Lipocalina 2/genética , Proteínas Asociadas a Pancreatitis/genética , beta-Defensinas/genética
4.
Gut ; 68(12): 2111-2121, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31563878

RESUMEN

Although faecal microbiota transplantation (FMT) has a well-established role in the treatment of recurrent Clostridioides difficile infection (CDI), its widespread dissemination is limited by several obstacles, including lack of dedicated centres, difficulties with donor recruitment and complexities related to regulation and safety monitoring. Given the considerable burden of CDI on global healthcare systems, FMT should be widely available to most centres.Stool banks may guarantee reliable, timely and equitable access to FMT for patients and a traceable workflow that ensures safety and quality of procedures. In this consensus project, FMT experts from Europe, North America and Australia gathered and released statements on the following issues related to the stool banking: general principles, objectives and organisation of the stool bank; selection and screening of donors; collection, preparation and storage of faeces; services and clients; registries, monitoring of outcomes and ethical issues; and the evolving role of FMT in clinical practice,Consensus on each statement was achieved through a Delphi process and then in a plenary face-to-face meeting. For each key issue, the best available evidence was assessed, with the aim of providing guidance for the development of stool banks in order to promote accessibility to FMT in clinical practice.


Asunto(s)
Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/terapia , Consenso , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Infecciones por Clostridium/microbiología , Selección de Donante , Humanos , Manejo de Especímenes/métodos
5.
BMC Microbiol ; 19(1): 167, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31319790

RESUMEN

BACKGROUND: The human gastrointestinal (GI) tract microbiota has been a subject of intense research throughout the 3rd Millennium. Now that a general picture about microbiota composition in health and disease is emerging, questions about factors determining development of microbiotas with specific community structures will be addressed. To this end, usage of murine models for colonization studies remains crucial. Optical in vivo imaging of either bioluminescent or fluorescent bacteria is the basis for non-invasive detection of intestinal colonization of bacteria. Although recent advances in in vivo fluorescence imaging have overcome many limitations encountered in bioluminescent imaging of intestinal bacteria, such as requirement for live cells, high signal attenuation and 2D imaging, the method is still restricted to bacteria for which molecular cloning tools are available. RESULTS: Here, we present usage of a lipophilic fluorescent dye together with Katushka far-red fluorescent protein to establish a dual-color in vivo imaging system to monitor GI transit of different bacterial strains, suitable also for strains resistant to genetic labeling. Using this system, we were able to distinguish two different E. coli strains simultaneously and show their unique transit patterns. Combined with fluorescence molecular tomography, these distinct strains could be spatially and temporally resolved and quantified in 3D. CONCLUSIONS: Developed novel method for labeling microbes and identify their passage both temporally and spatially in vivo makes now possible to monitor all culturable bacterial strains, also those that are resistant to conventional genetic labeling.


Asunto(s)
Tracto Gastrointestinal/microbiología , Microscopía Fluorescente/métodos , Coloración y Etiquetado/métodos , Animales , Escherichia coli/metabolismo , Colorantes Fluorescentes/metabolismo , Microbioma Gastrointestinal , Microscopía Intravital/métodos , Proteínas Luminiscentes/metabolismo , Lípidos de la Membrana/metabolismo , Ratones , Tomografía Óptica , Proteína Fluorescente Roja
6.
Gut ; 66(4): 569-580, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28087657

RESUMEN

Faecal microbiota transplantation (FMT) is an important therapeutic option for Clostridium difficile infection. Promising findings suggest that FMT may play a role also in the management of other disorders associated with the alteration of gut microbiota. Although the health community is assessing FMT with renewed interest and patients are becoming more aware, there are technical and logistical issues in establishing such a non-standardised treatment into the clinical practice with safety and proper governance. In view of this, an evidence-based recommendation is needed to drive the practical implementation of FMT. In this European Consensus Conference, 28 experts from 10 countries collaborated, in separate working groups and through an evidence-based process, to provide statements on the following key issues: FMT indications; donor selection; preparation of faecal material; clinical management and faecal delivery and basic requirements for implementing an FMT centre. Statements developed by each working group were evaluated and voted by all members, first through an electronic Delphi process, and then in a plenary consensus conference. The recommendations were released according to best available evidence, in order to act as guidance for physicians who plan to implement FMT, aiming at supporting the broad availability of the procedure, discussing other issues relevant to FMT and promoting future clinical research in the area of gut microbiota manipulation. This consensus report strongly recommends the implementation of FMT centres for the treatment of C. difficile infection as well as traces the guidelines of technicality, regulatory, administrative and laboratory requirements.


Asunto(s)
Clostridioides difficile , Enterocolitis Seudomembranosa/terapia , Trasplante de Microbiota Fecal , Selección de Paciente , Manejo de Especímenes/métodos , Selección de Donante , Europa (Continente) , Medicina Basada en la Evidencia , Trasplante de Microbiota Fecal/efectos adversos , Trasplante de Microbiota Fecal/métodos , Trasplante de Microbiota Fecal/normas , Instituciones de Salud , Unidades Hospitalarias/organización & administración , Humanos
7.
BMC Med ; 14(1): 155, 2016 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-27724956

RESUMEN

BACKGROUND: Faecal microbiota transplantation (FMT) is an effective treatment for recurrent Clostridium difficile infection (rCDI). It restores the disrupted intestinal microbiota and subsequently suppresses C. difficile. The long-term stability of the intestinal microbiota and the recovery of mucosal microbiota, both of which have not been previously studied, are assessed herein. Further, the specific bacteria behind the treatment efficacy are also investigated. METHODS: We performed a high-throughput microbiota profiling using a phylogenetic microarray analysis of 131 faecal and mucosal samples from 14 rCDI patients pre- and post-FMT during a 1-year follow-up and 23 samples from the three universal donors over the same period. RESULTS: The FMT treatment was successful in all patients. FMT reverted the patients' bacterial community to become dominated by Clostridium clusters IV and XIVa, the major anaerobic bacterial groups of the healthy gut. In the mucosa, the amount of facultative anaerobes decreased, whereas Bacteroidetes increased. Post-FMT, the patients' microbiota profiles were more similar to their own donors than what is generally observed for unrelated subjects and this striking similarity was retained throughout the 1-year follow-up. Furthermore, the universal donor approach allowed us to identify bacteria commonly established in all CDI patients and revealed a commonly acquired core microbiota consisting of 24 bacterial taxa. CONCLUSIONS: FMT induces profound microbiota changes, therefore explaining the high clinical efficacy for rCDI. The identification of commonly acquired bacteria could lead to effective bacteriotherapeutic formulations. FMT can affect microbiota in the long-term and offers a means to modify it relatively permanently for the treatment of microbiota-associated diseases.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Heces/microbiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Recurrencia , Resultado del Tratamiento
8.
Appl Environ Microbiol ; 82(19): 5756-62, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27422834

RESUMEN

UNLABELLED: Vancomycin-resistant enterococci (VRE) have become a major nosocomial threat. Enterococcus faecium is of special concern, as it can easily acquire new antibiotic resistances and is an excellent colonizer of the human intestinal tract. Several clinical studies have explored the potential use of beneficial bacteria to weed out opportunistic pathogens. Specifically, the widely studied Lactobacillus rhamnosus strain GG has been applied successfully in the context of VRE infections. Here, we provide new insight into the molecular mechanism underlying the effects of this model probiotic on VRE decolonization. Both clinical VRE isolates and L. rhamnosus GG express pili on their cell walls, which are the key modulators of their highly efficient colonization of the intestinal mucosa. We found that one of the VRE pilus clusters shares considerable sequence similarity with the SpaCBA-SrtC1 pilus cluster of L. rhamnosus GG. Remarkable immunological and functional similarities were discovered between the mucus-binding pili of L. rhamnosus GG and those of the clinical E. faecium strain E1165, which was characterized at the genome level. Moreover, E. faecium strain E1165 bound efficiently to mucus, which may be prevented by the presence of the mucus-binding SpaC protein or antibodies against L. rhamnosus GG or SpaC. These results present experimental support for a novel probiotic mechanism, in which the mucus-binding pili of L. rhamnosus GG prevent the binding of a potential pathogen to the host. Hence, we provide a molecular basis for the further exploitation of L. rhamnosus GG and its pilins for prophylaxis and treatment of VRE infections. IMPORTANCE: Concern about vancomycin-resistant Enterococcus faecium causing nosocomial infections is rising globally. The arsenal of antibiotic strategies to treat these infections is nearly exhausted, and hence, new treatment strategies are urgently needed. Here, we provide molecular evidence to underpin reports of the successful clinical application of Lactobacillus rhamnosus GG in VRE decolonization strategies. Our results provide support for a new molecular mechanism, in which probiotics can perform competitive exclusion and possibly immune interaction. Moreover, we spur further exploration of the potential of intact L. rhamnosus GG and purified SpaC pilin as prophylactic and curative agents of the VRE carrier state.


Asunto(s)
Enterococcus faecium/fisiología , Fimbrias Bacterianas/metabolismo , Lacticaseibacillus rhamnosus/fisiología , Interacciones Microbianas , Moco/microbiología , Probióticos/metabolismo , Humanos
9.
PLoS Genet ; 9(8): e1003683, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23966868

RESUMEN

Lactobacillus rhamnosus is a lactic acid bacterium that is found in a large variety of ecological habitats, including artisanal and industrial dairy products, the oral cavity, intestinal tract or vagina. To gain insights into the genetic complexity and ecological versatility of the species L. rhamnosus, we examined the genomes and phenotypes of 100 L. rhamnosus strains isolated from diverse sources. The genomes of 100 L. rhamnosus strains were mapped onto the L. rhamnosus GG reference genome. These strains were phenotypically characterized for a wide range of metabolic, antagonistic, signalling and functional properties. Phylogenomic analysis showed multiple groupings of the species that could partly be associated with their ecological niches. We identified 17 highly variable regions that encode functions related to lifestyle, i.e. carbohydrate transport and metabolism, production of mucus-binding pili, bile salt resistance, prophages and CRISPR adaptive immunity. Integration of the phenotypic and genomic data revealed that some L. rhamnosus strains possibly resided in multiple niches, illustrating the dynamics of bacterial habitats. The present study showed two distinctive geno-phenotypes in the L. rhamnosus species. The geno-phenotype A suggests an adaptation to stable nutrient-rich niches, i.e. milk-derivative products, reflected by the alteration or loss of biological functions associated with antimicrobial activity spectrum, stress resistance, adaptability and fitness to a distinctive range of habitats. In contrast, the geno-phenotype B displays adequate traits to a variable environment, such as the intestinal tract, in terms of nutrient resources, bacterial population density and host effects.


Asunto(s)
Genoma Bacteriano , Lacticaseibacillus rhamnosus/genética , Filogenia , Animales , Estudios de Asociación Genética , Genómica , Lacticaseibacillus rhamnosus/clasificación , Leche/microbiología , Fenotipo , Densidad de Población
10.
Anaerobe ; 39: 60-7, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26946362

RESUMEN

In our previous studies on the intestinal microbiota in irritable bowel syndrome (IBS), we identified a bacterial phylotype with higher abundance in patients suffering from diarrhea than in healthy controls. In the present work, we have isolated in pure culture strain RT94, belonging to this phylotype, determined its whole genome sequence and performed an extensive genomic analysis and phenotypical testing. This revealed strain RT94 to be a strict anaerobe apparently belonging to a novel species with only 94% similarity in the 16S rRNA gene sequence to the closest relatives Ruminococcus torques and Ruminococcus lactaris. The G + C content of strain RT94 is 45.2 mol% and the major long-chain cellular fatty acids are C16:0, C18:0 and C14:0. The isolate is metabolically versatile but not a mucus or cellulose utilizer. It produces acetate, ethanol, succinate, lactate and formate, but very little butyrate, as end products of glucose metabolism. The mechanisms underlying the association of strain RT94 with diarrhea-type IBS are discussed.


Asunto(s)
Bacterias Anaerobias/aislamiento & purificación , Diarrea/diagnóstico , Genoma Bacteriano , Infecciones por Bacterias Grampositivas/diagnóstico , Síndrome del Colon Irritable/diagnóstico , ARN Ribosómico 16S/genética , Ruminococcus/aislamiento & purificación , Ácido Acético/metabolismo , Bacterias Anaerobias/clasificación , Bacterias Anaerobias/genética , Composición de Base , Secuencia de Bases , Diarrea/microbiología , Etanol/metabolismo , Ácidos Grasos/metabolismo , Formiatos/metabolismo , Tracto Gastrointestinal/microbiología , Glucosa/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Síndrome del Colon Irritable/microbiología , Ácido Láctico/metabolismo , Filogenia , Ruminococcus/clasificación , Ruminococcus/genética , Análisis de Secuencia de ADN , Ácido Succínico/metabolismo
11.
Appl Environ Microbiol ; 81(11): 3655-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25795669

RESUMEN

Akkermansia muciniphila is a Gram-negative mucin-degrading bacterium that resides in the gastrointestinal tracts of humans and animals. A. muciniphila has been linked with intestinal health and improved metabolic status in obese and type 2 diabetic subjects. Specifically, A. muciniphila has been shown to reduce high-fat-diet-induced endotoxemia, which develops as a result of an impaired gut barrier. Despite the accumulating evidence of the health-promoting effects of A. muciniphila, the mechanisms of interaction of the bacterium with the host have received little attention. In this study, we used several in vitro models to investigate the adhesion of A. muciniphila to the intestinal epithelium and its interaction with the host mucosa. We found that A. muciniphila adheres strongly to the Caco-2 and HT-29 human colonic cell lines but not to human colonic mucus. In addition, A. muciniphila showed binding to the extracellular matrix protein laminin but not to collagen I or IV, fibronectin, or fetuin. Importantly, A. muciniphila improved enterocyte monolayer integrity, as shown by a significant increase in the transepithelial electrical resistance (TER) of cocultures of Caco-2 cells with the bacterium. Further, A. muciniphila induced interleukin 8 (IL-8) production by enterocytes at cell concentrations 100-fold higher than those for Escherichia coli, suggesting a very low level of proinflammatory activity in the epithelium. In conclusion, our results demonstrate that A. muciniphila adheres to the intestinal epithelium and strengthens enterocyte monolayer integrity in vitro, suggesting an ability to fortify an impaired gut barrier. These results support earlier associative in vivo studies and provide insights into the interaction of A. muciniphila with the host.


Asunto(s)
Adhesión Bacteriana , Enterocitos/microbiología , Células Epiteliales/fisiología , Verrucomicrobia/fisiología , Línea Celular , Enterocitos/inmunología , Enterocitos/metabolismo , Humanos , Interleucina-8/metabolismo , Verrucomicrobia/inmunología
12.
BMC Microbiol ; 15: 4, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25591990

RESUMEN

BACKGROUND: For a good probiotic candidate, the abilities to adhere to intestinal epithelium and to fortify barrier function are considered to be crucial for colonization and functionality of the strain. The strain Lactobacillus acidophilus LAB20 was isolated from the jejunum of a healthy dog, where it was found to be the most pre-dominant lactobacilli. In this study, the adhesion ability of LAB20 to intestinal epithelial cell (IECs) lines, IECs isolated from canine intestinal biopsies, and to canine, porcine and human intestinal mucus was investigated. Further, we studied the ability of LAB20 to fortify the epithelial cell monolayer and to reduce LPS-induced interleukin (IL-8) release from enterocytes. RESULTS: We found that LAB20 presented higher adhesion to canine colonic mucus as compared to mucus isolated from porcine colon. LAB20 showed adhesion to HT-29 and Caco-2 cell lines, and importantly also to canine IECs isolated from canine intestinal biopsies. In addition, LAB20 increased the transepithelial electrical resistance (TER) of enterocyte monolayers and thus strengthened the intestinal barrier function. The strain showed also anti-inflammatory capacity in being able to attenuate the LPS-induced IL-8 production of HT-29 cells. CONCLUSION: In conclusion, canine indigenous strain LAB20 is a potential probiotic candidate for dogs adhering to the host epithelium and showing intestinal barrier fortifying and anti-inflammatory effects.


Asunto(s)
Adhesión Bacteriana , Enterocitos/inmunología , Enterocitos/microbiología , Interleucina-8/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Lactobacillus acidophilus/fisiología , Animales , Células Cultivadas , Perros , Humanos , Interleucina-8/antagonistas & inhibidores , Yeyuno/microbiología , Lactobacillus acidophilus/aislamiento & purificación , Moco/metabolismo , Moco/microbiología
13.
Pediatr Res ; 77(4): 528-35, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25580735

RESUMEN

BACKGROUND: Bacterial contact in utero modulates fetal and neonatal immune responses. Maternal probiotic supplementation reduces the risk of immune-mediated disease in the infant. We investigated the immunomodulatory properties of live Lactobacillus rhamnosus GG and its SpaC pilus adhesin in human fetal intestinal models. METHODS: Tumor necrosis factor (TNF)-α mRNA expression was measured by qPCR in a human fetal intestinal organ culture model exposed to live L. rhamnosus GG and proinflammatory stimuli. Binding of recombinant SpaC pilus protein to intestinal epithelial cells (IECs) was assessed in human fetal intestinal organ culture and the human fetal intestinal epithelial cell line H4 by immunohistochemistry and immunofluorescence, respectively. TLR-related gene expression in fetal ileal organ culture after exposure to recombinant SpaC was assessed by qPCR. RESULTS: Live L. rhamnosus GG significantly attenuates pathogen-induced TNF-α mRNA expression in the human fetal gut. Recombinant SpaC protein was found to adhere to the fetal gut and to modulate varying levels of TLR-related gene expression. CONCLUSION: The human fetal gut is responsive to luminal microbes. L. rhamnosus GG significantly attenuates fetal intestinal inflammatory responses to pathogenic bacteria. The L. rhamnosus GG pilus adhesin SpaC binds to immature human IECs and directly modulates IEC innate immune gene expression.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Íleon/embriología , Íleon/microbiología , Lacticaseibacillus rhamnosus/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Toll-Like/metabolismo , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Citocinas/metabolismo , Células Epiteliales/citología , Fimbrias Bacterianas , Humanos , Inmunohistoquímica , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-10/metabolismo , Microscopía Fluorescente , Probióticos , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Tipo I de Interleucina-1/metabolismo , Proteínas Recombinantes/metabolismo , Salmonella typhimurium , Factor de Necrosis Tumoral alfa/metabolismo
14.
Scand J Gastroenterol ; 50(1): 34-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25523554

RESUMEN

Inflammatory bowel diseases (IBDs) are chronic debilitating disorders of unknown etiology, consisting of two main conditions, ulcerative colitis and Crohn's disease. Major advances have recently taken place in human genetic studies of IBD and over 160 risk loci for these two diseases have been uncovered. These genetic data highlight a key role for genes that code for immunological and epithelial barrier functions. Environmental factors also make substantial contributions to the pathogenesis of IBD and account for the growing incidence of the diseases around the world. Intestinal microbiota creates resistance to infection, provides nutrients, and educates the immune system and in many ways has a significant impact on human health. Aberrant microbiota composition and decreased diversity (dysbiotic microbiota) are key etiopathological events in IBD. Dysbiotic microbiota can lead to loss of normal, regulatory immune effects in the gut mucosa. This may play a central role in the development and perpetuation of chronic inflammation. Further, the expression of specific innate immune receptors that recognize microbes is altered in the IBD epithelium. Therefore, the combination of host side epithelial barrier functions and the presence of dysbiotic microbiota in the gut together promote inflammation. New therapeutic options targeting microbiota are currently considered for IBD and they may, in the future, provide means to reverse the pathogenic host-microbiota relationship into a symbiotic one. In this review, the focus is on the intestinal microbiota and host-microbe interactions in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/microbiología , Microbiota , Homeostasis/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/terapia , Probióticos/uso terapéutico
15.
Curr Top Microbiol Immunol ; 358: 323-46, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22094893

RESUMEN

Our intestinal tract is colonized since birth by multiple microbial species that show a characteristic succession in time. Notably the establishment of the microbiota in early life is important as it appears to impact later health. While apparently stable in healthy adults, the intestinal microbiota is changing significantly during aging. After 100 years of symbiosis marked changes have been observed that may relate to an increased level of intestinal inflammation. There is considerable interest in the microbiota in health and disease as it may provide functional biomarkers, the possibility to differentiate subjects, and avenues for interventions. This chapter reviews the present state of the art on the research to investigate the contribution of the intestinal microbiota to human health. Specific attention will be given to the healthy microbiota and aberrations due to disturbances such as celiac disease, irritable bowel syndrome, inflammatory bowel disease, obesity and diabetes, and non-alcoholic fatty liver disease.


Asunto(s)
Bacterias/metabolismo , Enfermedad/historia , Salud/historia , Intestinos/microbiología , Metagenoma , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Mucosa Intestinal/metabolismo
16.
JAMA Surg ; 159(6): 606-614, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38506889

RESUMEN

Importance: Surgical site infections (SSIs)-especially anastomotic dehiscence-are major contributors to morbidity and mortality after rectal resection. The role of mechanical and oral antibiotics bowel preparation (MOABP) in preventing complications of rectal resection is currently disputed. Objective: To assess whether MOABP reduces overall complications and SSIs after elective rectal resection compared with mechanical bowel preparation (MBP) plus placebo. Design, Setting, and Participants: This multicenter, double-blind, placebo-controlled randomized clinical trial was conducted at 3 university hospitals in Finland between March 18, 2020, and October 10, 2022. Patients aged 18 years and older undergoing elective resection with primary anastomosis of a rectal tumor 15 cm or less from the anal verge on magnetic resonance imaging were eligible for inclusion. Outcomes were analyzed using a modified intention-to-treat principle, which included all patients who were randomly allocated to and underwent elective rectal resection with an anastomosis. Interventions: Patients were stratified according to tumor distance from the anal verge and neoadjuvant treatment given and randomized in a 1:1 ratio to receive MOABP with an oral regimen of neomycin and metronidazole (n = 277) or MBP plus matching placebo tablets (n = 288). All study medications were taken the day before surgery, and all patients received intravenous antibiotics approximately 30 minutes before surgery. Main Outcomes and Measures: The primary outcome was overall cumulative postoperative complications measured using the Comprehensive Complication Index. Key secondary outcomes were SSI and anastomotic dehiscence within 30 days after surgery. Results: In all, 565 patients were included in the analysis, with 288 in the MBP plus placebo group (median [IQR] age, 69 [62-74] years; 190 males [66.0%]) and 277 in the MOABP group (median [IQR] age, 70 [62-75] years; 158 males [57.0%]). Patients in the MOABP group experienced fewer overall postoperative complications (median [IQR] Comprehensive Complication Index, 0 [0-8.66] vs 8.66 [0-20.92]; Wilcoxon effect size, 0.146; P < .001), fewer SSIs (23 patients [8.3%] vs 48 patients [16.7%]; odds ratio, 0.45 [95% CI, 0.27-0.77]), and fewer anastomotic dehiscences (16 patients [5.8%] vs 39 patients [13.5%]; odds ratio, 0.39 [95% CI, 0.21-0.72]) compared with patients in the MBP plus placebo group. Conclusions and Relevance: Findings of this randomized clinical trial indicate that MOABP reduced overall postoperative complications as well as rates of SSIs and anastomotic dehiscences in patients undergoing elective rectal resection compared with MBP plus placebo. Based on these findings, MOABP should be considered as standard treatment in patients undergoing elective rectal resection. Trial Registration: ClinicalTrials.gov Identifier: NCT04281667.


Asunto(s)
Antibacterianos , Neoplasias del Recto , Infección de la Herida Quirúrgica , Humanos , Masculino , Femenino , Método Doble Ciego , Persona de Mediana Edad , Infección de la Herida Quirúrgica/prevención & control , Infección de la Herida Quirúrgica/epidemiología , Anciano , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Neoplasias del Recto/cirugía , Administración Oral , Profilaxis Antibiótica , Cuidados Preoperatorios/métodos , Neomicina/administración & dosificación , Neomicina/uso terapéutico , Catárticos/administración & dosificación , Metronidazol/administración & dosificación , Metronidazol/uso terapéutico , Proctectomía/efectos adversos , Recto/cirugía , Dehiscencia de la Herida Operatoria/prevención & control , Dehiscencia de la Herida Operatoria/etiología , Procedimientos Quirúrgicos Electivos/efectos adversos
17.
Gut Microbes ; 16(1): 2295445, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38214604

RESUMEN

Altered microbiota and impaired host immune function have been linked to the pathogenesis of pouchitis. We used 16S rRNA gene sequencing and RNA sequencing data from a previous randomized clinical trial (RCT) on fecal microbiota transplantation (FMT) therapy in 26 chronic pouchitis patients with one-year follow-up. We analyzed changes in both luminal and mucosal microbiota composition, as well as in host mucosal gene expression to gain insights into the host-microbiota interactions possibly underlying clinical outcomes of the patients. Antibiotic type and pattern of use were significant drivers of the luminal microbiota at baseline. Differential gene expression analysis indicated transition from ileal to colonic gene expression in the pouch, and upregulation in inflammation- and immune system-related pathways in the pouch. At 4 weeks, the non-relapsed FMT patients had a lower microbiota dissimilarity to the donor than the non-relapsed placebo patients (p = .02). While two FMT-treated patients showed a shift toward the donor's microbiota during the one-year follow-up, the overall FMT microbiota modulation effect was low. Patient's luminal and mucosal microbiota profiles were unstable in both FMT and placebo groups. Expression of the chemokine receptor CXCR4 was downregulated at 52 weeks compared to the baseline in the non-relapsed patients in both FMT and placebo groups. Microbiota modulation by FMT seems to be low in this patient group. The microbiota composition or alterations did not explain the relapse status of the patients. Some evidence for remission-related host gene expression pattern was found; specifically, CXCR4 expression may have a role in sustained remission.


Asunto(s)
Colitis Ulcerosa , Microbioma Gastrointestinal , Microbiota , Reservoritis , Humanos , Trasplante de Microbiota Fecal/efectos adversos , Reservoritis/terapia , Reservoritis/metabolismo , Colitis Ulcerosa/terapia , Expresión Génica , Heces
18.
Appl Environ Microbiol ; 79(22): 6989-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24014530

RESUMEN

The ability of bifidobacteria to adhere to the intestine of the human host is considered to be important for efficient colonization and achieving probiotic effects. Bifidobacterium bifidum strains DSM20456 and MIMBb75 adhere well to the human intestinal cell lines Caco-2 and HT-29. The surface lipoprotein BopA was previously described to be involved in mediating adherence of B. bifidum to epithelial cells, but thioacylated, purified BopA inhibited the adhesion of B. bifidum to epithelial cells in competitive adhesion assays only at very high concentrations, indicating an unspecific effect. In this study, the role of BopA in the adhesion of B. bifidum was readdressed. The gene encoding BopA was cloned and expressed without its lipobox and hydrophobic signal peptide in Escherichia coli, and an antiserum against the recombinant BopA was produced. The antiserum was used to demonstrate the abundant localization of BopA on the cell surface of B. bifidum. However, blocking of B. bifidum BopA with specific antiserum did not reduce adhesion of bacteria to epithelial cell lines, arguing that BopA is not an adhesin. Also, adhesion of B. bifidum to human colonic mucin and fibronectin was found to be BopA independent. The recombinant BopA bound only moderately to human epithelial cells and colonic mucus, and it failed to bind to fibronectin. Thus, our results contrast the earlier findings on the major role of BopA in adhesion, indicating that the strong adhesion of B. bifidum to epithelial cell lines is BopA independent.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Bifidobacterium/fisiología , Células Epiteliales/microbiología , Mucosa Intestinal/citología , Proteínas de la Membrana/metabolismo , Proteínas Bacterianas/genética , Bifidobacterium/genética , Células CACO-2 , Membrana Celular , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Genes Bacterianos , Células HT29 , Humanos , Mucosa Intestinal/microbiología , Lipoproteínas/genética , Lipoproteínas/metabolismo , Proteínas de la Membrana/genética , Moco/microbiología , Probióticos , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Appl Environ Microbiol ; 79(6): 1923-33, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23315726

RESUMEN

Four Lactobacillus strains were isolated from marketed probiotic products, including L. rhamnosus strains from Vifit (Friesland Campina) and Idoform (Ferrosan) and L. casei strains from Actimel (Danone) and Yakult (Yakult Honsa Co.). Their genomes and phenotypes were characterized and compared in detail with L. casei strain BL23 and L. rhamnosus strain GG. Phenotypic analysis of the new isolates indicated differences in carbohydrate utilization between L. casei and L. rhamnosus strains, which could be linked to their genotypes. The two isolated L. rhamnosus strains had genomes that were virtually identical to that of L. rhamnosus GG, testifying to their genomic stability and integrity in food products. The L. casei strains showed much greater genomic heterogeneity. Remarkably, all strains contained an intact spaCBA pilus gene cluster. However, only the L. rhamnosus strains produced mucus-binding SpaCBA pili under the conditions tested. Transcription initiation mapping demonstrated that the insertion of an iso-IS30 element upstream of the pilus gene cluster in L. rhamnosus strains but absent in L. casei strains had constituted a functional promoter driving pilus gene expression. All L. rhamnosus strains triggered an NF-κB response via Toll-like receptor 2 (TLR2) in a reporter cell line, whereas the L. casei strains did not or did so to a much lesser extent. This study demonstrates that the two L. rhamnosus strains isolated from probiotic products are virtually identical to L. rhamnosus GG and further highlights the differences between these and L. casei strains widely marketed as probiotics, in terms of genome content, mucus-binding and metabolic capacities, and host signaling capabilities.


Asunto(s)
Lacticaseibacillus casei/genética , Lacticaseibacillus casei/fisiología , Lacticaseibacillus rhamnosus/genética , Lacticaseibacillus rhamnosus/fisiología , Probióticos , Técnicas de Tipificación Bacteriana , Variación Genética , Genotipo , Lacticaseibacillus casei/inmunología , Lacticaseibacillus casei/metabolismo , Lacticaseibacillus rhamnosus/inmunología , Lacticaseibacillus rhamnosus/metabolismo , Fenotipo
20.
BMC Microbiol ; 13: 12, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23339708

RESUMEN

BACKGROUND: Deviations in composition and diversity of intestinal microbiota in infancy have been associated with both the development and recurrence of atopic eczema. Thus, we decided to use a deep and global microarray-based method to characterize the diversity and temporal changes of the intestinal microbiota in infancy and to define specific bacterial signatures associated with eczema. Faecal microbiota at 6 and 18 months of age were analysed from 34 infants (15 with eczema and 19 healthy controls) selected from a prospective follow-up study based on the availability of faecal samples. The infants were originally randomized to receive either Lactobacillus rhamnosus GG or placebo. RESULTS: Children with eczema harboured a more diverse total microbiota than control subjects as assessed by the Simpson's reciprocal diversity index of the microarray profiles. Composition of the microbiota did not differ between study groups at age of 6 months, but was significantly different at age of 18 months as assessed by MCPP (p=0.01). At this age healthy children harboured 3 -fold greater amount of members of the Bacteroidetes (p=0.01). Microbiota of children suffering from eczema had increased abundance of the Clostridium clusters IV and XIVa, which are typically abundant in adults. Probiotic Lactobacillus rhamnosus GG supplementation in early infancy was observed to have minor long-term effects on the microbiota composition. CONCLUSION: A diverse and adult-type microbiota in early childhood is associated with eczema and it may contribute to the perpetuation of eczema.


Asunto(s)
Bacterias/clasificación , Bacterias/genética , Biodiversidad , Dermatitis Atópica/microbiología , Tracto Gastrointestinal/microbiología , Metagenoma , Análisis por Micromatrices , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Placebos/administración & dosificación , Probióticos/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA