Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
mBio ; 12(2)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785619

RESUMEN

Clostridioides difficile, a Gram-positive, spore-forming bacterium, is the primary cause of infectious nosocomial diarrhea. Antibiotics are a major risk factor for C. difficile infection (CDI), as they disrupt the gut microbial community, enabling increased germination of spores and growth of vegetative C. difficile To date, the only single-species bacterial preparation that has demonstrated efficacy in reducing recurrent CDI in humans is nontoxigenic C. difficile Using multiple infection models, we determined that precolonization with a less virulent strain is sufficient to protect from challenge with a lethal strain of C. difficile, surprisingly even in the absence of adaptive immunity. Additionally, we showed that protection is dependent on high levels of colonization by the less virulent strain and that it is mediated by exclusion of the invading strain. Our results suggest that reduction of amino acids, specifically glycine following colonization by the first strain of C. difficile, is sufficient to decrease germination of the second strain, thereby limiting colonization by the lethal strain.IMPORTANCE Antibiotic-associated colitis is often caused by infection with the bacterium Clostridioides difficile In this study, we found that reduction of the amino acid glycine by precolonization with a less virulent strain of C. difficile is sufficient to decrease germination of a second strain. This finding demonstrates that the axis of competition for nutrients can include multiple life stages. This work is important, as it is the first to identify a possible mechanism through which precolonization with C. difficile, a current clinical therapy, provides protection from reinfection. Furthermore, our work suggests that targeting nutrients utilized by all life stages could be an improved strategy for bacterial therapeutics that aim to restore colonization resistance in the gut.


Asunto(s)
Antibiosis , Terapia Biológica , Clostridioides difficile/fisiología , Infecciones por Clostridium/prevención & control , Animales , Clostridioides difficile/clasificación , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Femenino , Glicina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota , Esporas Bacterianas/clasificación , Esporas Bacterianas/genética , Esporas Bacterianas/crecimiento & desarrollo , Esporas Bacterianas/fisiología , Virulencia
2.
Kaohsiung J Med Sci ; 35(6): 327-331, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31017741

RESUMEN

Fecal microbiota transplantation (FMT) had been an ancient remedy for severe illness several centuries ago. Under modern medical analysis and evidence-based research, it has been proved as an alternative treatment for recurrent Clostridium difficile infection and recent randomized control study also showed that FMT could be an adjuvant treatment for inflammatory bowel disease. As we get a better understanding of the relationship between gut microbiota and systemic disease, FMT became a potential treatment to explore. This article summarized procedures such as donor selection, fecal material preparation, transplantation delivery methods, and adverse events. We also review the present evidence about FMT in clinical practice.


Asunto(s)
Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal/métodos , Enfermedades Inflamatorias del Intestino/terapia , Administración Oral , Administración Rectal , Clostridioides difficile/patogenicidad , Clostridioides difficile/fisiología , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Estreñimiento/etiología , Estreñimiento/fisiopatología , Diarrea/etiología , Diarrea/fisiopatología , Enema/métodos , Trasplante de Microbiota Fecal/efectos adversos , Heces/microbiología , Fiebre/etiología , Fiebre/fisiopatología , Flatulencia/etiología , Flatulencia/fisiopatología , Microbioma Gastrointestinal/fisiología , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Vómitos/etiología , Vómitos/fisiopatología
3.
mSphere ; 4(2)2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894434

RESUMEN

Between October 2016 and June 2017, a C57BL/6J mouse colony that was undergoing a pre- and perinatal methyl donor supplementation diet intervention to study the impact of parental nutrition on offspring susceptibility to disease was found to suffer from an epizootic of unexpected deaths. Necropsy revealed the presence of severe colitis, and further investigation linked these outbreak deaths to a Clostridium difficile strain of ribotype 027 that we term 16N203. C. difficile infection (CDI) is associated with antibiotic use in humans. Current murine models of CDI rely on antibiotic pretreatment to establish clinical phenotypes. In this report, the C. difficile outbreak occurs in F1 mice linked to alterations in the parental diet. The diagnosis of CDI in the affected mice was confirmed by cecal/colonic histopathology, the presence of C. difficile bacteria in fecal/colonic culture, and detection of C. difficile toxins. F1 mice from parents fed the methyl supplementation diet also had significantly reduced survival (P < 0.0001) compared with F1 mice from parents fed the control diet. When we tested the 16N203 outbreak strain in an established mouse model of antibiotic-induced CDI, we confirmed that this strain is pathogenic. Our serendipitous observations from this spontaneous outbreak of C. difficile in association with a pre- and perinatal methyl donor diet suggest the important role that diet may play in host defense and CDI risk factors.IMPORTANCEClostridium difficile infection (CDI) has become the leading cause of infectious diarrhea in hospitals worldwide, owing its preeminence to the emergence of hyperendemic strains, such as ribotype 027 (RT027). A major CDI risk factor is antibiotic exposure, which alters gut microbiota, resulting in the loss of colonization resistance. Current murine models of CDI also depend on pretreatment of animals with antibiotics to establish disease. The outbreak that we report here is unique in that the CDI occurred in mice with no antibiotic exposure and is associated with a pre- and perinatal methyl supplementation donor diet intervention study. Our investigation subsequently reveals that the outbreak strain that we term 16N203 is an RT027 strain, and this isolated strain is also pathogenic in an established murine model of CDI (with antibiotics). Our report of this spontaneous outbreak offers additional insight into the importance of environmental factors, such as diet, and CDI susceptibility.


Asunto(s)
Infecciones por Clostridium/etiología , Dieta/efectos adversos , Suplementos Dietéticos/efectos adversos , Brotes de Enfermedades , Animales , Betaína/metabolismo , Colina/metabolismo , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/patogenicidad , Susceptibilidad a Enfermedades/etiología , Femenino , Masculino , Metionina/metabolismo , Ratones , Ratones Endogámicos C57BL , Nutrición Parenteral/métodos , Ribotipificación , Factores de Riesgo
5.
Artículo en Inglés | MEDLINE | ID: mdl-29439962

RESUMEN

DS-2969b is a novel GyrB inhibitor that is currently under clinical development for the treatment of Clostridium difficile infection (CDI). In this study, the in vitro and in vivo activities of DS-2969b were evaluated. DS-2969b inhibited the supercoiling activity of C. difficile DNA gyrase. DS-2969b showed potent in vitro activity against C. difficile clinical isolates with a MIC90 of 0.06 µg/ml, which was 2-, 32-, and 16-fold lower than the MIC90s of fidaxomicin, vancomycin, and metronidazole, respectively. DS-2969b did not select spontaneously resistant mutants of various C. difficile strains at 4× MIC, and the frequency of resistance development was less than 4.8 × 10-9 In a hamster CDI model, 5-day oral administration of DS-2969b conferred complete protection from recurrence and mortality at 0.3 mg/kg of body weight once a day, in contrast to a 50% survival rate with fidaxomicin at 3 mg/kg once a day and 0% with vancomycin at a 50-mg/kg/dose twice a day. Even a single oral administration of 1 mg/kg of DS-2969b in the CDI model exhibited 100% animal survival without recurrence. DS-2969b was also efficacious by 5-day subcutaneous administration in the CDI model. DS-2969b showed similar levels of fecal excretion after intravenous and oral administrations in rats. These data support further development of DS-2969b as a drug for oral and intravenous treatment of CDI.


Asunto(s)
Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Administración Oral , Animales , Antibacterianos/administración & dosificación , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Masculino , Mesocricetus , Metronidazol/administración & dosificación , Metronidazol/uso terapéutico , Pruebas de Sensibilidad Microbiana , Ratas , Ratas Sprague-Dawley , Vancomicina/administración & dosificación , Vancomicina/uso terapéutico
6.
JPEN J Parenter Enteral Nutr ; 42(7): 1156-1167, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29385239

RESUMEN

BACKGROUND: Clostridium difficile (CD) infection (CDI) increases patient morbidity, mortality and healthcare costs. Antibiotic treatment induces gut dysbiosis and is both a major risk factor for CD colonization and treatment of CDI. Probiotics have been trialed to support commensal gut microbiota and reduce CDI. This study investigated commensal microbe Faecalibacterium prausnitzii (FP) and a prebiotic, both known to yield butyrate and be anti-inflammatory and immunomodulatory, on CD colonization and gut integrity in mice. METHODS: Mice were randomly grouped and supplemented daily with FP, prebiotic, FP + prebiotic, FP/prebiotic supernatant, or saline throughout the entire study. Following treatment with clindamycin for 3 days, mice were exposed to CD. Feces were collected at baseline, the day after antibiotic, and 1, 3, and 5 days after CD exposure and cultured for bacterial overgrowth and CD colonization. On days 1 and 5 after CD exposure, mice were randomly euthanized, and proximal colon was dissected for histological analysis and preparation of RNA for analysis of proinflammatory and anti-inflammatory cytokines. RESULTS: Although all mice exhibited bacterial overgrowth and CD colonization, bacterial burden resolved quicker in the FP + prebiotic group. This was associated with induction and resolution of innate immune responses, anion exchanger, and tight junction protein preservation in proximal colon. CD toxin virulence potential was questionable as expression of CD toxin B receptor was depleted in the FP + prebiotic group. CONCLUSION: Supplementation with anti-inflammatory butyrate-supporting commensal bacteria and prebiotic may support innate immune responses and minimize bacterial burden and negative effects during antibiotic and CD exposure.


Asunto(s)
Antibacterianos/efectos adversos , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/tratamiento farmacológico , Faecalibacterium prausnitzii , Microbioma Gastrointestinal , Prebióticos , Probióticos/uso terapéutico , Animales , Proteínas de Transporte de Anión/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Butiratos/metabolismo , Butiratos/farmacología , Clindamicina/efectos adversos , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/microbiología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Disbiosis/etiología , Faecalibacterium prausnitzii/crecimiento & desarrollo , Faecalibacterium prausnitzii/metabolismo , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Ratas Sprague-Dawley , Receptores Inmunológicos/metabolismo , Solanum tuberosum/química , Almidón/farmacología , Almidón/uso terapéutico , Proteínas de Uniones Estrechas/metabolismo
7.
APMIS ; 125(11): 1033-1038, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28960474

RESUMEN

Anaerobic bacteria, such as Bacteroides fragilis or Clostridium perfringens, are part of indigenous human flora. However, Clostridium difficile represents also an important causative agent of nosocomial infectious antibiotic-associated diarrhoea. Treatment of C. difficile infection is problematic, making it imperative to search for new compounds with antimicrobial properties. Hops (Humulus lupulus L.) contain substances with antibacterial properties. We tested antimicrobial activity of purified hop constituents humulone, lupulone and xanthohumol against anaerobic bacteria. The antimicrobial activity was established against B. fragilis, C. perfringens and C. difficile strains according to standard testing protocols (CLSI, EUCAST), and the minimum inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBC) were calculated. All C. difficile strains were toxigenic and clinically relevant, as they were isolated from patients with diarrhoea. Strongest antimicrobial effects were observed with xanthohumol showing MIC and MBC values of 15-107 µg/mL, which are close to those of conventional antibiotics in the strains of bacteria with increased resistance. Slightly higher MIC and MBC values were obtained with lupulone followed by higher values of humulone. Our study, thus, shows a potential of purified hop compounds, especially xanthohumol, as alternatives for treatment of infections caused by select anaerobic bacteria, namely nosocomial diarrhoea caused by resistant strains.


Asunto(s)
Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Ciclohexenos/farmacología , Flavonoides/farmacología , Humulus/química , Propiofenonas/farmacología , Terpenos/farmacología , Anaerobiosis/fisiología , Antibacterianos/aislamiento & purificación , Bacteroides fragilis/efectos de los fármacos , Bacteroides fragilis/crecimiento & desarrollo , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Clostridium perfringens/efectos de los fármacos , Clostridium perfringens/crecimiento & desarrollo , Infección Hospitalaria/microbiología , Ciclohexenos/aislamiento & purificación , Diarrea/microbiología , Enterocolitis Seudomembranosa/microbiología , Flavonoides/aislamiento & purificación , Humanos , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/química , Propiofenonas/aislamiento & purificación , Simbiosis/fisiología , Terpenos/aislamiento & purificación
8.
Benef Microbes ; 8(2): 143-151, 2017 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-28008787

RESUMEN

The probiotic definition requires the administration of an 'adequate amount' in order to obtain a health benefit. What that amount should be is not indicated. Here, an overview is given of studies that investigated the dose-response relation of probiotics in human interventions. Studies were divided in; meta-analyses, meta-analyses on specific probiotic strains, and studies testing two or more doses of a probiotic (combination) in the same study. Meta-analyses on the effect of probiotics on antibiotic associated diarrhoea (AAD) suggest a dose-response effect; for Clostridium difficile-associated diarrhoea on the other hand no dose-response was observed. For other end-points; such as necrotising enterocolitis, prevention of atopic dermatitis and slow intestinal transit, no dose-response relation was identified in meta-analyses. For prophylaxis in colorectal cancer and relief of irritable bowel syndrome, no dose-response relation was determined. However, for blood pressure, a meta-analysis observed that higher doses (greater than 1011 cfu) were more effective than lower doses. Meta-analyses of specific strains suggest a break-point for effectiveness of Lactobacillus rhamnosus GG in the treatment of acute gastroenteritis in children; no dose-response was observed for two other probiotics assessed. Studies comparing two or more doses indicate that faecal recovery and risk reduction of AAD follow a positive dose-response relationship. Other end-points such as immune markers, general health, and bowel function did not exhibit clear dose-response relations. For AAD, the findings are very compelling; both meta-analyses and dedicated dose-response studies observe a positive correlation between dose and AAD risk. These findings do not allow for extrapolation, but suggest that studying higher doses for this end-point would be worthwhile. The lack of a clear dose-response for other end-points, does not mean it does not exist; present data does just not allow drawing any conclusions.


Asunto(s)
Antibacterianos/uso terapéutico , Bifidobacterium , Enterocolitis Seudomembranosa/terapia , Gastroenteritis/terapia , Lacticaseibacillus rhamnosus , Probióticos/uso terapéutico , Saccharomyces boulardii , Clostridioides difficile/patogenicidad , Suplementos Dietéticos/microbiología , Relación Dosis-Respuesta a Droga , Humanos
9.
PLoS One ; 11(1): e0147210, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26789728

RESUMEN

Fecal microbiota transplantation (FMT) is a highly effective therapy for recurrent Clostridium difficile infection (R-CDI), but its mechanisms remain poorly understood. Emerging evidence suggests that gut bile acids have significant influence on the physiology of C. difficile, and therefore on patient susceptibility to recurrent infection. We analyzed spore germination of 10 clinical C. difficile isolates exposed to combinations of bile acids present in patient feces before and after FMT. Bile acids at concentrations found in patients' feces prior to FMT induced germination of C. difficile, although with variable potency across different strains. However, bile acids at concentrations found in patients after FMT did not induce germination and inhibited vegetative growth of all C. difficile strains. Sequencing of the newly identified germinant receptor in C. difficile, CspC, revealed a possible correspondence of variation in germination responses across isolates with mutations in this receptor. This may be related to interstrain variability in spore germination and vegetative growth in response to bile acids seen in this and other studies. These results support the idea that intra-colonic bile acids play a key mechanistic role in the success of FMT, and suggests that novel therapeutic alternatives for treatment of R-CDI may be developed by targeted manipulation of bile acid composition in the colon.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Terapia Biológica/métodos , Clostridioides difficile/crecimiento & desarrollo , Colon/metabolismo , Enterocolitis Seudomembranosa/prevención & control , Trasplante de Microbiota Fecal , Heces/microbiología , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/patogenicidad , Colon/microbiología , Enterocolitis Seudomembranosa/microbiología , Humanos
10.
Int Rev Immunol ; 35(1): 67-82, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25970037

RESUMEN

In space, the lifestyle, relative sterility of spaceship and extreme environmental stresses, such as microgravity and cosmic radiation, can compromise the balance between human body and human microbiome. An astronaut's body during spaceflight encounters increased risk for microbial infections and conditions because of immune dysregulation and altered microbiome, i.e. dysbiosis. This risk is further heightened by increase in virulence of pathogens in microgravity. Health status of astronauts might potentially benefit from maintaining a healthy microbiome by specifically managing their diet on space in addition to probiotic therapies. This review focuses on the current knowledge/understanding of how spaceflight affects human immunity and microbiome.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad/efectos de la radiación , Vuelo Espacial , Ingravidez/efectos adversos , Astronautas , Bacteroides/inmunología , Bacteroides/efectos de la radiación , Candida albicans/inmunología , Candida albicans/patogenicidad , Clostridiales/inmunología , Clostridiales/patogenicidad , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Radiación Cósmica/efectos adversos , Citocinas/inmunología , Citocinas/metabolismo , Citocinas/efectos de la radiación , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Suplementos Dietéticos , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Microbioma Gastrointestinal/efectos de la radiación , Helicobacter hepaticus/inmunología , Helicobacter hepaticus/patogenicidad , Humanos , Leucocitos/metabolismo , Leucocitos/efectos de la radiación , Probióticos/uso terapéutico , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/patogenicidad , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Virulencia
11.
J Food Sci ; 80(12): M2925-31, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26556797

RESUMEN

Clostridium difficile infection (CDI) is the most prevalent cause of health-care-associated infections. CDI-related health-care costs and deaths are both increasing annually on a global scale. C. difficile have been reported in food products in Canada, Europe, and the United States; however, the systematic transmission of C. difficile between humans and animals is yet to be understood. Because of the limitations of current therapeutic options, there is a need for the development of new patient treatments. Epigallocatechin gallate (EGCG) is a major catechin compound found in green tea extracts and exhibits antioxidant and antimicrobial activities. This study was conducted to investigate the inhibitory effects of EGCG on the expression of virulence genes in C. difficile and in C. difficile-associated diseases by inhibition of quorum sensing. The protein expression of autoinducer-2 (AI-2) was evaluated by AI-2 activity. EGCG at various concentrations had an inhibitory effect on AI-2 production, especially at 10 µg/mL. EGCG also significantly repressed the transcription of virulence genes, including luxS and tcdA, and prolonged the survival of Caenorhabditis elegans infected with C. difficile. Furthermore, treatment with EGCG effectively protected C. difficile-infected mice from C. difficile-induced death. Histological analysis of the colon and cecum of these mice revealed that EGCG protected tissues of the lower intestinal tract from damage. EGCG exerted growth-inhibitory and bactericidal activities on C. difficile in C. difficile-infected mice. Our results suggest that EGCG has significant antipathogenic effects on C. difficile and can be used to prevent or treat C. difficile-associated diseases or C. difficile infections.


Asunto(s)
Antibacterianos/uso terapéutico , Camellia sinensis/química , Catequina/análogos & derivados , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , Fitoterapia , Virulencia/efectos de los fármacos , Animales , Antibacterianos/farmacología , Caenorhabditis elegans , Catequina/farmacología , Catequina/uso terapéutico , Clostridioides difficile/genética , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Expresión Génica/efectos de los fármacos , Genes Bacterianos , Homoserina/análogos & derivados , Homoserina/metabolismo , Lactonas/metabolismo , Ratones , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Reacción en Cadena de la Polimerasa , Percepción de Quorum , Ribotipificación , Especificidad de la Especie , Virulencia/genética
12.
J Clin Microbiol ; 53(11): 3702-4, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26354812

RESUMEN

Large clostridial toxin-negative, binary toxin-positive (A(-) B(-) CDT(+)) strains of Clostridium difficile are almost never associated with clinically significant C. difficile infection (CDI), possibly because such strains are not detected by most diagnostic methods. We report the isolation of an A(-) B(-) CDT(+) ribotype 033 (RT033) strain of C. difficile from a young patient with ulcerative colitis and severe diarrhea.


Asunto(s)
ADP Ribosa Transferasas/genética , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Clostridioides difficile/genética , Enterocolitis Seudomembranosa/tratamiento farmacológico , Enterotoxinas/genética , ADP Ribosa Transferasas/metabolismo , Adolescente , Australia , Proteínas Bacterianas/metabolismo , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/patogenicidad , Colitis Ulcerosa/microbiología , Enterocolitis Seudomembranosa/microbiología , Heces/microbiología , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Recurrencia
13.
Gastroenterology ; 149(1): 223-37, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25982290

RESUMEN

The community of microorganisms within the human gut (or microbiota) is critical to health and functions with a level of complexity comparable to that of an organ system. Alterations of this ecology (or dysbiosis) have been implicated in a number of disease states, and the prototypical example is Clostridium difficile infection (CDI). Fecal microbiota transplantation (FMT) has been demonstrated to durably alter the gut microbiota of the recipient and has shown efficacy in the treatment of patients with recurrent CDI. There is hope that FMT may eventually prove beneficial for the treatment of other diseases associated with alterations in gut microbiota, such as inflammatory bowel disease, irritable bowel syndrome, and metabolic syndrome, to name a few. Although the basic principles that underlie the mechanisms by which FMT shows therapeutic efficacy in CDI are becoming apparent, further research is needed to understand the possible role of FMT in these other conditions. Although relatively simple to perform, questions regarding both short-term and long-term safety as well as the complex and rapidly evolving regulatory landscape has limited widespread use. Future work will focus on establishing best practices and more robust safety data than exist currently, as well as refining FMT beyond current "whole-stool" transplants to increase safety and tolerability. Encapsulated formulations, full-spectrum stool-based products, and defined microbial consortia are all in the immediate future.


Asunto(s)
Terapia Biológica/métodos , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/terapia , Heces/microbiología , Microbiota , Adulto , Clostridioides difficile/aislamiento & purificación , Femenino , Humanos , Masculino , Persona de Mediana Edad
14.
Infect Control Hosp Epidemiol ; 36(6): 681-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25783204

RESUMEN

OBJECTIVE: To determine the potential epidemiologic and economic value of the implementation of a multifaceted Clostridium difficile infection (CDI) control program at US acute care hospitals DESIGN: Markov model with a 5-year time horizon PARTICIPANTS: Patients whose data were used in our simulations were limited to hospitalized Medicare beneficiaries ≥65 years old. BACKGROUND: CDI is an important public health problem with substantial associated morbidity, mortality, and cost. Multifaceted national prevention efforts in the United Kingdom, including antimicrobial stewardship, patient isolation, hand hygiene, environmental cleaning and disinfection, and audit, resulted in a 59% reduction in CDI cases reported from 2008 to 2012. METHODS: Our analysis was conducted from the federal perspective. The intervention we modeled included the following components: antimicrobial stewardship utilizing the Antimicrobial Use and Resistance module of the National Healthcare Safety Network (NHSN), use of contact precautions, and enhanced environmental cleaning. We parameterized our model using data from CDC surveillance systems, the AHRQ Healthcare Cost and Utilization Project, and literature reviews. To address uncertainty in our parameter estimates, we conducted sensitivity analyses for intervention effectiveness and cost, expenditures by other federal partners, and discount rate. Each simulation represented a cohort of 1,000 hospitalized patients over 1,000 trials. RESULTS In our base case scenario with 50% intervention effectiveness, we estimated that 509,000 CDI cases and 82,000 CDI-attributable deaths would be prevented over a 5-year time horizon. Nationally, the cost savings across all hospitalizations would be $2.5 billion (95% credible interval: $1.2 billion to $4.0 billion). CONCLUSIONS: The potential benefits of a multifaceted national CDI prevention program are sizeable from the federal perspective.


Asunto(s)
Antibacterianos , Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa , Control de Infecciones/métodos , Anciano , Anciano de 80 o más Años , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Análisis Costo-Beneficio , Infección Hospitalaria/prevención & control , Enterocolitis Seudomembranosa/epidemiología , Enterocolitis Seudomembranosa/etiología , Enterocolitis Seudomembranosa/prevención & control , Humanos , Control de Infecciones/economía , Cadenas de Markov , Administración del Tratamiento Farmacológico/estadística & datos numéricos , Programas Nacionales de Salud , Administración de la Seguridad/economía , Administración de la Seguridad/métodos , Estados Unidos/epidemiología
15.
World J Gastroenterol ; 21(1): 6-11, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25574076

RESUMEN

Fecal microbiota transplantation (FMT) has garnered significant attention in recent years in the face of a reemerging Clostridium difficile (C. difficile) epidemic. Positive results from the first randomized control trial evaluating FMT have encouraged the medical community to explore the process further and expand its application beyond C. difficile infections and even the gastrointestinal domain. However promising and numerous the prospects of FMT appear, the method remains limited in scope today due to several important barriers, most notably a poorly defined federal regulatory policy. The Food and Drug Administration has found it difficult to standardize and regulate the administration of inherently variable, metabolically active, and ubiquitously available fecal material. The current cumbersome policy, which classifies human feces as a drug, has prevented physicians from providing FMT and deserving patients from accessing FMT in a timely fashion, and subsequent modifications seem only to be temporary. The argument for reclassifying fecal material as human tissue is well supported. Essentially, this would allow for a regulatory framework that is sufficiently flexible to expand access to care and facilitate research, but also appropriately restrictive and centralized to ensure patient safety. Such an approach can facilitate the advancement of FMT to a more refined, controlled, and aesthetic process, perhaps in the form of a customized and well-characterized stool substitute therapy.


Asunto(s)
Terapia Biológica/métodos , Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa/terapia , Heces/microbiología , Política de Salud , Intestinos/microbiología , Microbiota , Animales , Terapia Biológica/efectos adversos , Enterocolitis Seudomembranosa/diagnóstico , Enterocolitis Seudomembranosa/epidemiología , Enterocolitis Seudomembranosa/microbiología , Humanos , Seguridad del Paciente , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento
16.
J Clin Gastroenterol ; 48 Suppl 1: S80-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25291136

RESUMEN

Gut microbiota is deeply involved in the regulation of both health and disease within our body. The restoration of a healthy gut microbiota is, therefore, a main clinical target in the management of diseases associated with its disruption. Fecal microbiota transplantation (FMT) is an old therapy that has recently been rediscovered, having proved a clear efficacy against recurrent Clostridium difficile infection. By restoring the altered gut microbiota in a substantial and durable manner, FMT is considered a cutting-edge promising option for the treatment of disease that recognize the alteration of the gut microbiota as having a pathogenic role. FMT has shown interesting (even if uncertain) results in diseases such as metabolic syndrome and inflammatory bowel diseases. Moreover, the definition of a standard procedural protocol for each specific disease, as well as exhaustive studies about the relationship between donor's microbiota composition and clinical results, will certainly improve the therapeutic potential of FMT. Both the application of cutting-edge technologies for the assessment of gut microbiota composition (such as metagenomics) and the development of well-designed, large randomized trials are needed to put such perspectives into practice.


Asunto(s)
Terapia Biológica/métodos , Heces/microbiología , Intestinos/microbiología , Animales , Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa/diagnóstico , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/terapia , Humanos , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/microbiología , Enfermedades del Sistema Inmune/terapia , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/terapia , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Sistema Nervioso/microbiología , Enfermedades del Sistema Nervioso/terapia , Resultado del Tratamiento
17.
World J Gastroenterol ; 20(35): 12687-90, 2014 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-25253977

RESUMEN

Acute respiratory distress syndrome is a life-threatening disorder caused mainly by pneumonia. Clostridium difficile infection (CDI) is a common nosocomial diarrheal disease. Disruption of normal intestinal flora by antibiotics is the main risk factor for CDI. The use of broad-spectrum antibiotics for serious medical conditions can make it difficult to treat CDI complicated by acute respiratory distress syndrome. Fecal microbiota transplantation is a highly effective treatment in patients with refractory CDI. Here we report on a patient with refractory CDI and acute respiratory distress syndrome caused by pneumonia who was treated with fecal microbiota transplantation.


Asunto(s)
Antibacterianos/efectos adversos , Terapia Biológica/métodos , Clostridioides difficile/patogenicidad , Colon/microbiología , Duodenoscopía , Enterocolitis Seudomembranosa/terapia , Heces/microbiología , Neumonía Bacteriana/terapia , Síndrome de Dificultad Respiratoria/terapia , Anciano de 80 o más Años , Enterocolitis Seudomembranosa/diagnóstico , Enterocolitis Seudomembranosa/microbiología , Humanos , Masculino , Neumonía Bacteriana/diagnóstico , Neumonía Bacteriana/microbiología , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/microbiología , Sigmoidoscopía , Factores de Tiempo , Resultado del Tratamiento
18.
Antimicrob Agents Chemother ; 58(10): 5714-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25022586

RESUMEN

SMT19969 [2,2'-bis(4-pyridyl)3H,3'-H 5,5-bibenzimidazole] is a novel narrow-spectrum nonabsorbable antibiotic currently in development for the treatment of Clostridium difficile infection. The comparative activities of SMT19969 and vancomycin against nonepidemic and epidemic strains of C. difficile were studied in an established hamster model. Against nonepidemic (VA11) strains, the survival rates of SMT19969-treated animals ranged from 80% to 95%. Vancomycin exhibited 100% protection during treatment, with relapse observed starting on day 9 and 50% survival at day 20. At 50 mg/kg of body weight, SMT19969 administered orally once daily for 5 days provided full protection of treated animals on the dosing days and through day 12 against epidemic strains. Vancomycin also protected during the dosing interval, but apparent relapse occurred earlier, starting on day 11. SMT19969 exhibited excellent in vitro activity, with MICs of 0.25 µg/ml for all isolates. The MICs for vancomycin were 2- to 4-fold higher at ≤0.5 to 1 µg/ml. All plasma sample concentrations of SMT19969 were below the limit of quantification (25 ng/ml) at all time points, consistent with the reported lack of bioavailability of the compound. Cecal concentrations were significantly above the MIC (ranging from 96 µg/ml to 172 µg/ml).


Asunto(s)
Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Animales , Infecciones por Clostridium/microbiología , Cricetinae , Masculino , Mesocricetus , Metronidazol/uso terapéutico , Pruebas de Sensibilidad Microbiana , Vancomicina/uso terapéutico
19.
World J Gastroenterol ; 20(23): 7416-23, 2014 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-24966611

RESUMEN

Clostridium difficile infections (CDI) are a leading cause of antibiotic-associated and nosocomial diarrhea. Despite effective antibiotic treatments, recurrent infections are common. With the recent emergence of hypervirulent isolates of C. difficile, CDI is a growing epidemic with higher rates of recurrence, increasing severity and mortality. Fecal microbiota transplantation (FMT) is an alternative treatment for recurrent CDI. A better understanding of intestinal microbiota and its role in CDI has opened the door to this promising therapeutic approach. FMT is thought to resolve dysbiosis by restoring gut microbiota diversity thereby breaking the cycle of recurrent CDI. Since the first reported use of FMT for recurrent CDI in 1958, systematic reviews of case series and case report have shown its effectiveness with high resolution rates compared to standard antibiotic treatment. This article focuses on current guidelines for CDI treatment, the role of intestinal microbiota in CDI recurrence and current evidence about FMT efficacy, adverse effects and acceptability.


Asunto(s)
Clostridioides difficile/patogenicidad , Enterocolitis Seudomembranosa/microbiología , Intestinos/microbiología , Microbiota , Antibacterianos/efectos adversos , Terapia Biológica/efectos adversos , Terapia Biológica/métodos , Enterocolitis Seudomembranosa/diagnóstico , Enterocolitis Seudomembranosa/terapia , Heces/microbiología , Humanos , Intestinos/efectos de los fármacos , Microbiota/efectos de los fármacos , Recurrencia , Factores de Riesgo , Resultado del Tratamiento , Virulencia
20.
Gastroenterology ; 146(6): 1547-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24503131

RESUMEN

Antibiotic-associated infection with the bacterial pathogen Clostridium difficile is a major cause of morbidity and increased health care costs. C difficile infection follows disruption of the indigenous gut microbiota by antibiotics. Antibiotics create an environment within the intestine that promotes C difficile spore germination, vegetative growth, and toxin production, leading to epithelial damage and colitis. Studies of patients with C difficile infection and animal models have shown that the indigenous microbiota can inhibit expansion and persistence of C difficile. Although the specific mechanisms of these processes are not known, they are likely to interfere with key aspects of the pathogen's physiology, including spore germination and competitive growth. Increasing our understanding of how the intestinal microbiota manage C difficile could lead to better means of controlling this important nosocomial pathogen.


Asunto(s)
Clostridioides difficile/crecimiento & desarrollo , Enterocolitis Seudomembranosa/microbiología , Intestinos/microbiología , Microbiota , Animales , Antibacterianos/efectos adversos , Ácidos y Sales Biliares/metabolismo , Terapia Biológica/métodos , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Modelos Animales de Enfermedad , Disbiosis , Enterocolitis Seudomembranosa/metabolismo , Enterocolitis Seudomembranosa/prevención & control , Heces/microbiología , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Microbiota/efectos de los fármacos , Probióticos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA