Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Curr Opin Clin Nutr Metab Care ; 25(5): 334-340, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35838278

RESUMEN

PURPOSE OF REVIEW: To provide an up-to-date review on the clinical assessment of two important gastrointestinal problems with overlapping symptomatology but diverse cause and testing methods. Small intestinal bacterial overgrowth (SIBO) is characterized by the presence of excess bacteria in the small intestine associated with bloating, distention, gas, and diarrhea. Lactose intolerance is caused by lactase enzyme deficiency in the small bowel mucosa leading to lactose malabsorption and symptoms of bloating, gas, and diarrhea. RECENT FINDINGS: SIBO is assessed by hydrogen/methane breath test using glucose as a substrate and/or small bowel aspirate and culture but these tests have shortcomings. Consequently, several new diagnostic techniques, including novel capsule technologies and other approaches are being evaluated. Lactose intolerance can be assessed by hydrogen/methane breath test using lactose as a substrate, or small bowel mucosal lactase assay, genetic testing and lactose tolerance test, although the efficacy and practicality of these diagnostic modalities are not equal. SUMMARY: In clinical practice, gas, bloating, distention, pain, and diarrhea are common gastrointestinal symptoms that often remain unexplained when routine gastrointestinal endoscopy, imaging, and stool tests are negative. These patients should be evaluated for SIBO and/or food intolerances including lactose intolerance.


Asunto(s)
Intolerancia a la Lactosa , Pruebas Respiratorias/métodos , Diarrea/diagnóstico , Diarrea/etiología , Humanos , Hidrógeno , Lactasa , Intolerancia a la Lactosa/diagnóstico , Intolerancia a la Lactosa/microbiología , Metano
2.
mBio ; 13(3): e0105122, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35695459

RESUMEN

An outstanding question regarding the human gut microbiota is whether and how microbiota-directed interventions influence host phenotypic traits. Here, we employed a dietary intervention to probe this question in the context of lactose intolerance. To assess the effects of dietary dairy product elimination and (re)introduction on the microbiota and host phenotype, we studied 12 self-reported mildly lactose-intolerant adults with triweekly collection of fecal samples over a 12-week study period: 2 weeks of baseline diet, 4 weeks of dairy product elimination, and 6 weeks of gradual whole cow milk (re)introduction. Of the 12 subjects, 6 reported either no dairy or only lactose-free dairy product consumption. A clinical assay for lactose intolerance, the hydrogen breath test, was performed before and after each of these three study phases, and 16S rRNA gene amplicon sequencing was performed on all fecal samples. We found that none of the subjects showed change in a clinically defined measure of lactose tolerance. Similarly, fecal microbiota structure resisted modification. Although the mean fraction of the genus Bifidobacterium, a group known to metabolize lactose, increased slightly with milk (re)introduction (from 0.0125 to 0.0206; Wilcoxon P = 0.068), the overall structure of each subject's gut microbiota remained highly individualized and largely stable in the face of diet manipulation. IMPORTANCE Lactose intolerance is a gastrointestinal disorder diagnosed with a lactose hydrogen breath test. Lifestyle changes such as diet interventions can impact the gut microbiome; however, the role of the microbiome in lactose intolerance is unclear. Our study assessed the effects of a 12-week dietary dairy product elimination and (re)introduction on the microbiome and clinical lactose intolerance status in 12 adult self-reported lactose-intolerant individuals. We found each subject's gut microbiome remained highly individualized and largely stable in the face of this diet manipulation. We also report that none of the subjects showed change in a clinically defined measure of lactose tolerance.


Asunto(s)
Microbioma Gastrointestinal , Intolerancia a la Lactosa , Animales , Bovinos , Femenino , Humanos , Hidrógeno/metabolismo , Lactosa/análisis , Lactosa/metabolismo , Intolerancia a la Lactosa/metabolismo , Intolerancia a la Lactosa/microbiología , Leche/química , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Autoinforme
3.
FP Essent ; 516: 31-37, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35507311

RESUMEN

Malabsorption syndromes are a heterogenous group of conditions that can cause distressing gastrointestinal symptoms. Celiac disease is most common and is triggered by exposure to gluten. Tissue transglutaminase immunoglobulin A is the diagnostic test of choice; management is gluten avoidance. Lactose intolerance is caused by absence or declining levels of the enzyme lactase. Diagnosis typically is clinical, but breath tests can be helpful if diagnosis is uncertain. Management is lactose avoidance. Bile acid malabsorption results in unabsorbed bile acids in the colon, leading to diarrhea. The 75selenium homotaurocholic acid test is most accurate but is not widely available. Therefore, a trial of bile acid sequestrants (typically cholestyramine) is a reasonable alternative when the diagnosis is suspected. Exocrine pancreatic insufficiency is caused by decreased production of pancreatic enzymes, typically occurring in patients with preexisting pancreatic damage from alcohol, surgery, radiation, diabetes, or cystic fibrosis. Diagnosis involves fecal fat or fecal elastase-1 tests. Management is pancreatic enzyme replacement. Small intestinal bacterial overgrowth is caused by pathologic overgrowth of the small bowel microbiome. Diagnosis is by jejunal biopsy or, more commonly, breath tests. Antibiotics (typically rifaximin) are the initial management. Other options include dietary changes, probiotics, and prokinetic drugs.


Asunto(s)
Enfermedad Celíaca , Intolerancia a la Lactosa , Síndromes de Malabsorción , Ácidos y Sales Biliares , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/terapia , Glútenes , Humanos , Intolerancia a la Lactosa/complicaciones , Intolerancia a la Lactosa/diagnóstico , Intolerancia a la Lactosa/microbiología , Síndromes de Malabsorción/diagnóstico , Síndromes de Malabsorción/etiología , Síndromes de Malabsorción/terapia
4.
Gut Microbes ; 13(1): 1957536, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34365905

RESUMEN

Background. Our recent publication (Chey et al., Nutrients 2020) showed that a 30-day administration of pure galacto-oligosaccharides (GOS) significantly reduced symptoms and altered the fecal microbiome in patients with lactose intolerance (LI). Results. In this addendum, we performed an in-depth analysis of the fecal microbiome of the 377 LI patients randomized to one of two GOS doses (Low, 10-15 grams/day or High, 15-20 grams/day), or placebo in a multi-center, double-blinded, placebo-controlled trial. Sequencing of 16S rRNA amplicons was done on GOS or placebo groups at weeks zero (baseline), four (end of treatment), nine, 16 and 22. Taxa impacted by treatment and subsequent dairy consumption included lactose-fermenting species of Bifidobacterium, Lactobacillus, Lactococcus, and Streptococcus. Increased secondary fermentation microorganisms included Coprococcus and Ruminococcus species, Blautia producta, and Methanobrevibacterium. Finally, tertiary fermenters that use acetate to generate butyrate were also increased, including Faecalibacterium prausnitzii, Roseburia faecis, and C. eutactus. Conclusions. Results confirmed and expanded data on GOS microbiome modulation in LI individuals. Microbiome analysis at 16 and 22 weeks after treatment further suggested relatively long-term benefits when individuals continued consumption of dairy products.


Asunto(s)
Actinobacteria/aislamiento & purificación , Clostridiales/aislamiento & purificación , Microbioma Gastrointestinal/fisiología , Intolerancia a la Lactosa/microbiología , Oligosacáridos/metabolismo , Prebióticos/administración & dosificación , Actinobacteria/clasificación , Actinobacteria/genética , Clostridiales/clasificación , Clostridiales/genética , Método Doble Ciego , Humanos , Lactobacillus/crecimiento & desarrollo , Lactobacillus/metabolismo , Oligosacáridos/administración & dosificación , Placebos/administración & dosificación , ARN Ribosómico 16S/genética
5.
PLoS One ; 15(5): e0230784, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32384113

RESUMEN

BACKGROUND: Lactose intolerance is a frequent gastrointestinal disease affecting 47% of the Eastern European population. Small intestinal bacterial overgrowth (SIBO) leads to carbohydrate malabsorption and therefore to false results during lactose breath and tolerance tests. OBJECTIVES: We aimed to assess the prevalence of lactose maldigestion and intolerance in Hungary and to investigate the role of combined diagnostic method and testing for SIBO in reducing false results. METHODS: We retrospectively analyzed data from 264 adult symptomatic patients who underwent 50g lactose breath and tolerance tests in parallel over a one-year period at our center. A ≥20 ppm elevation of H2 or less than 1.1 mmol/l rise of blood glucose was diagnostic for lactose maldigestion. Patients with maldigestion who had symptoms during the test were defined as lactose intolerant. Patients with an early (≤90 min) significant (≥20 ppm) rise of H2 during lactose and/or lactulose breath tests were determined to have SIBO. Patients with slow/rapid oro-cecal transit and inappropriate preparation before the test were excluded. RESULTS: 49.6% of the 264 patients had lactose maldigestion, and 29.5% had lactose intolerance. The most frequent symptom was bloating (22.7%), while 34.8% of the study population and 60% of the symptomatic patients had SIBO. In 9.1% and 9.8% of the patients, the lactose breath and tolerance test alone gave false positive result compared with the combined method. SIBO was present in 75% of the false positives diagnosed with breath test only. CONCLUSIONS: The prevalence of lactose intolerance is lower in Hungary compared to the Eastern European value (29.5% vs 47%), so it is worth performing a population-based prospective analysis in this area. A combination of lactose breath and tolerance tests and the careful monitoring of results (with early H2 rise, lactulose breath test, etc.) can decrease the false cases caused by e.g. SIBO.


Asunto(s)
Infecciones Bacterianas/diagnóstico , Intestino Delgado/microbiología , Síndrome del Colon Irritable/diagnóstico , Intolerancia a la Lactosa/diagnóstico , Adulto , Infecciones Bacterianas/epidemiología , Infecciones Bacterianas/microbiología , Infecciones Bacterianas/patología , Pruebas Respiratorias , Reacciones Falso Positivas , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Hungría/epidemiología , Hidrógeno , Síndrome del Colon Irritable/epidemiología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/patología , Lactosa/administración & dosificación , Intolerancia a la Lactosa/epidemiología , Intolerancia a la Lactosa/microbiología , Intolerancia a la Lactosa/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos
6.
Nutrients ; 12(4)2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290344

RESUMEN

Background and Aims: Lactose intolerance (LI) is a global problem affecting more than half of the world's population. An ultra-purified, high-concentration galacto-oligosaccharide, RP-G28, is being developed as a treatment for patients with LI. The efficacy and safety of RP-G28 in reducing symptoms of lactose intolerance were assessed in a blinded, randomized, placebo-controlled trial. Methods: In this multiclinical site, double-blinded, placebo-controlled trial, 377 patients with LI were randomized to one of two doses of orally administered RP-G28 or placebo for 30 days. A LI test and symptom assessment were performed at baseline and on day 31. The primary endpoint was a ≥4-point reduction or a score of zero on LI composite score on day 31. Voluntary milk and dairy intake and global outcome measures assessed patients' overall treatment satisfaction and quality of life before therapy and 30 days after therapy. This study received Institutional Review Board (IRB) approval. Results: For the primary endpoint, 40% in the RP-G28 groups reported a ≥4-point reduction or no symptoms on LI symptom composite score compared to 26% with placebo (P = 0.016). Treatment with RP-G28 also led to significantly higher levels of milk and dairy intake and significant improvements in global assessments compared to placebo. RP-G28 but not placebo led to significant increases in five Bifidobacterium taxa. Conclusions: RP-G28 for 30 days significantly reduced symptoms and altered the fecal microbiome in patients with LI. Treatment with RP-G28 also improved milk/dairy consumption and quality of life and was safe and well tolerated.


Asunto(s)
Intolerancia a la Lactosa/tratamiento farmacológico , Oligosacáridos/administración & dosificación , Administración Oral , Adolescente , Adulto , Anciano , Bifidobacterium , Productos Lácteos , Método Doble Ciego , Ingestión de Alimentos , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Intolerancia a la Lactosa/microbiología , Masculino , Persona de Mediana Edad , Calidad de Vida , Seguridad , Resultado del Tratamiento , Adulto Joven
7.
Nutrients ; 11(4)2019 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-31010241

RESUMEN

Functional gastrointestinal symptoms are frequent, and may be driven by several pathogenic mechanisms. Symptoms may persist in lactose intolerant (LI) patients (i.e., subjects with intestinal lactase deficiency, lactose malabsorption producing symptoms), after a lactose-free diet. Our hypothesis was that probiotic and vitamin B6 treatment may be useful to alleviate symptoms in LI patients through a positive modulation of gut microbial composition and relative metabolism. We aimed to test the efficacy of a novel formulation of Bifidobacterium longum BB536 and Lactobacillus rhamnosus HN001 plus vitamin B6 (ZR) in 23 LI subjects with persistent symptoms during a lactose-free diet. Symptoms, microbiome, and metabolome were measured at baseline and after 30 days in a crossover, randomized, double-blind study of ZR versus placebo (PL). Compared with PL, the administration of probiotics and vitamin B6 significantly decreased bloating (p = 0.028) and ameliorated constipation (p = 0.045). Fecal microbiome differed between ZR and PL. ZR drove the enrichment of several genera involved in lactose digestion including Bifidobacerium. Moreover, the relative abundance of acetic acid, 2-methyl-propanoic acid, nonenal, and indolizine 3-methyl increased, while phenol decreased. Our findings highlight the importance of selected probiotics and vitamin B6 to alleviate symptoms and gut dysbiosis in lactose intolerant patients with persistent functional gastrointestinal symptoms.


Asunto(s)
Bifidobacterium longum/crecimiento & desarrollo , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Intolerancia a la Lactosa/tratamiento farmacológico , Lactosa/metabolismo , Probióticos/uso terapéutico , Adulto , Estreñimiento/etiología , Estreñimiento/prevención & control , Estudios Cruzados , Dieta , Método Doble Ciego , Disbiosis/prevención & control , Heces/microbiología , Humanos , Intestinos , Lactasa/deficiencia , Intolerancia a la Lactosa/complicaciones , Intolerancia a la Lactosa/metabolismo , Intolerancia a la Lactosa/microbiología , Masculino , Persona de Mediana Edad , Vitamina B 6/farmacología , Vitamina B 6/uso terapéutico , Complejo Vitamínico B/farmacología , Complejo Vitamínico B/uso terapéutico
8.
Nutrients ; 10(10)2018 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-30279333

RESUMEN

The aim of this study was to compare the impact of whole milk supplementation on gut microbiota and cardiometabolic biomarkers between lactose malabsorbers (LM) and absorbers (LA). We performed a pair-wise intervention study of 31 LM and 31 LA, 1:1 matched by age, sex, body mass index, and daily dairy intake. Subjects were required to add 250 mL/day whole milk for four weeks in their routine diet. At the beginning and the end of the intervention period, we collected data on gut microbiota and cardiometabolic biomarkers. Whole milk supplementation significantly increased Actinobacteria (P < 0.01), Bifidobacterium (P < 0.01), Anaerostipe (P < 0.01), and Blautia (P = 0.04), and decreased Megamonas (P = 0.04) in LM, but not LA. Microbial richness and diversity were not affected. The fecal levels of short-chain fatty acids (SCFAs) remained stable throughout the study. Body fat mass (P < 0.01) and body fat percentage (P < 0.01) reduced in both groups, but the changes did not differ between groups. No significant differences in other cardiometabolic markers were found between LM and LA. When compared with LA, whole milk supplementation could alter the intestinal microbiota composition in LM, without significant changes in fecal SCFAs and cardiometabolic biomarkers.


Asunto(s)
Dieta/métodos , Microbioma Gastrointestinal/fisiología , Intolerancia a la Lactosa/sangre , Intolerancia a la Lactosa/microbiología , Leche/metabolismo , Actinobacteria/aislamiento & purificación , Adulto , Animales , Bifidobacterium/aislamiento & purificación , Biomarcadores/sangre , Heces/microbiología , Femenino , Humanos , Masculino , Leche/química , Adulto Joven
9.
Nutrients ; 10(10)2018 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332787

RESUMEN

Lactose intolerance, characterized by a decrease in host lactase expression, affects approximately 75% of the world population. Galacto-oligosaccharides (GOS) are prebiotics that have been shown to alleviate symptoms of lactose intolerance and to modulate the intestinal microbiota, promoting the growth of beneficial microorganisms. We hypothesized that mechanisms of GOS utilization by intestinal bacteria are variable, impacting efficacy and response, with differences occurring at the strain level. This study aimed to determine the mechanisms by which human-derived Lactobacillus rhamnosus strains metabolize GOS. Genomic comparisons between strains revealed differences in carbohydrate utilization components, including transporters, enzymes for degradation, and transcriptional regulation, despite a high overall sequence identity (>95%) between strains. Physiological and transcriptomics analyses showed distinct differences in carbohydrate metabolism profiles and GOS utilization between strains. A putative operon responsible for GOS utilization was identified and characterized by genetic disruption of the 6-phospho-ß-galactosidase, which had a critical role in GOS utilization. Our findings highlight the importance of strain-specific bacterial metabolism in the selection of probiotics and synbiotics to alleviate symptoms of gastrointestinal disorders including lactose intolerance.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus/metabolismo , Intolerancia a la Lactosa/microbiología , Oligosacáridos/metabolismo , Prebióticos , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Humanos , Lacticaseibacillus rhamnosus/genética
10.
Annu Rev Genet ; 51: 413-433, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-28934590

RESUMEN

The body's microbiome, composed of microbial cells that number in the trillions, is involved in human health and disease in ways that are just starting to emerge. The microbiome is assembled at birth, develops with its host, and is greatly influenced by environmental factors such as diet and other exposures. Recently, a role for human genetic variation has emerged as also influential in accounting for interpersonal differences in microbiomes. Thus, human genes may influence health directly or by promoting a beneficial microbiome. Studies of the heritability of gut microbiotas reveal a subset of microbes whose abundances are partly genetically determined by the host. However, the use of genome-wide association studies (GWASs) to identify human genetic variants associated with microbiome phenotypes has proven challenging. Studies to date are small by GWAS standards, and cross-study comparisons are hampered by differences in analytical approaches. Nevertheless, associations between microbes or microbial genes and human genes have emerged that are consistent between human populations. Most notably, higher levels of beneficial gut bacteria called Bifidobacteria are associated with the human lactase nonpersister genotype, which typically confers lactose intolerance, in several different human populations. It is time for the microbiome to be incorporated into studies that quantify interactions among genotype, environment, and the microbiome in order to predict human disease susceptibility.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Microbioma Gastrointestinal/fisiología , Genoma Humano , Intolerancia a la Lactosa/genética , Obesidad/genética , Esquizofrenia/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/microbiología , Esclerosis Amiotrófica Lateral/patología , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Dieta/métodos , Tracto Gastrointestinal/microbiología , Variación Genética , Estudio de Asociación del Genoma Completo , Genotipo , Genética Humana , Humanos , Intolerancia a la Lactosa/metabolismo , Intolerancia a la Lactosa/microbiología , Intolerancia a la Lactosa/patología , Obesidad/metabolismo , Obesidad/microbiología , Obesidad/patología , Fenotipo , Carácter Cuantitativo Heredable , Esquizofrenia/metabolismo , Esquizofrenia/microbiología , Esquizofrenia/patología
11.
Nutrients ; 7(9): 8020-35, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26393648

RESUMEN

Lactose intolerance related to primary or secondary lactase deficiency is characterized by abdominal pain and distension, borborygmi, flatus, and diarrhea induced by lactose in dairy products. The biological mechanism and lactose malabsorption is established and several investigations are available, including genetic, endoscopic and physiological tests. Lactose intolerance depends not only on the expression of lactase but also on the dose of lactose, intestinal flora, gastrointestinal motility, small intestinal bacterial overgrowth and sensitivity of the gastrointestinal tract to the generation of gas and other fermentation products of lactose digestion. Treatment of lactose intolerance can include lactose-reduced diet and enzyme replacement. This is effective if symptoms are only related to dairy products; however, lactose intolerance can be part of a wider intolerance to variably absorbed, fermentable oligo-, di-, monosaccharides and polyols (FODMAPs). This is present in at least half of patients with irritable bowel syndrome (IBS) and this group requires not only restriction of lactose intake but also a low FODMAP diet to improve gastrointestinal complaints. The long-term effects of a dairy-free, low FODMAPs diet on nutritional health and the fecal microbiome are not well defined. This review summarizes recent advances in our understanding of the genetic basis, biological mechanism, diagnosis and dietary management of lactose intolerance.


Asunto(s)
Productos Lácteos/efectos adversos , Dieta Baja en Carbohidratos , Tracto Gastrointestinal , Lactasa/deficiencia , Intolerancia a la Lactosa/dietoterapia , Lactosa/efectos adversos , Adulto , Digestión , Fermentación , Absorción Gastrointestinal , Microbioma Gastrointestinal , Tracto Gastrointestinal/enzimología , Tracto Gastrointestinal/microbiología , Predisposición Genética a la Enfermedad , Humanos , Lactasa/genética , Lactasa/inmunología , Lactasa/metabolismo , Lactosa/metabolismo , Intolerancia a la Lactosa/diagnóstico , Intolerancia a la Lactosa/genética , Intolerancia a la Lactosa/inmunología , Intolerancia a la Lactosa/microbiología , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Resultado del Tratamiento
12.
Ter Arkh ; 87(2): 20-23, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-25864343

RESUMEN

AIM: To establish the rate of lactase deficiency (LD) in patients with post-infectious irritable bowel syndrome (PI-IBS), to define a role of enteric bacteria in the pathogenesis of hypolactasia, and to evaluate the efficiency of probiotic therapy. SUBJECTS AND METHODS: Examinations were made in 386 patients with PI-IBS, including 112 (79.4%) women; mean age 33.9 ± 9.1 years; disease duration 2.6 ± 1.4 years. Rapid tests of small intestinal mucosa (SIM) biopsy specimens obtained from the duodenal retrobulbar segment were used to diagnose LD. Bacterial growth was estimated by a hydrogen breath test using a H2 MICRO gas analyzer. RESULTS: The patients with PI-IBS were revealed to have moderate and severe LD in 25.6 and 10.9%, respectively. All the patients with LD were detected to have small intestinal (SI) bacterial overgrowth (BOG). An inverse correlation was found between LD and the degree of SI BOG (r = -0.53; p < 0.001). 73.7% of the patients with moderate LD showed a positive effect of probiotic therapy as regression of clinical symptoms of LD, a decrease of hydrogen levels in expired air from 72.4 ± 25.1 to 16.41 ± 3.2 ppm (p < 0.05), an increase of lactate activity in the SIM biopsy specimens and an improvement of quality of life from 2.69 ± 0.53 to 5.53 ± 0.64 scores according to the GCI scale. No improvement occurred in 73.8% of the patients with severe LD. CONCLUSION: LD was identified in 36.5% of the patients with PI-IBS. There was an inverse correlation between the degree of LD and SI BOG. The good therapeutic effect of probiotics in LD suggests that the symbiotic gut microflora positively affects the activity of lactase in the human SIM. No therapeutic effect of probiotics in patients with severe LN serves as the basis for a search for more active probiotic therapy.


Asunto(s)
Intestino Delgado/microbiología , Síndrome del Colon Irritable/microbiología , Lactasa/deficiencia , Intolerancia a la Lactosa/microbiología , Probióticos/uso terapéutico , Adulto , Femenino , Humanos , Intestino Delgado/enzimología , Síndrome del Colon Irritable/dietoterapia , Síndrome del Colon Irritable/enzimología , Intolerancia a la Lactosa/dietoterapia , Intolerancia a la Lactosa/enzimología , Masculino , Resultado del Tratamiento
13.
J Gastroenterol Hepatol ; 30(6): 995-1000, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25470082

RESUMEN

BACKGROUND AND AIM: It has been reported that small intestinal bacterial overgrowth (SIBO) may lead to false positive diagnoses of lactose malabsorption (LM) in irritable bowel syndrome patients. The aim of this study was to determine the influence of SIBO on lactose hydrogen breath test (HBT) results in these patients. METHODS: Diarrhea-predominant irritable bowel syndrome patients with abnormal lactose HBTs ingested a test meal containing (99m) Tc and lactose. The location of the test meal and the breath levels of hydrogen were recorded simultaneously by scintigraphic scanning and lactose HBT, respectively. The increase in hydrogen concentration was not considered to be caused by SIBO if ≥ 10% of (99m) Tc accumulated in the cecal region at the time or before of abnormal lactose HBT. RESULTS: LM was present in 84% (31/37) of irritable bowel syndrome patients. Twenty of these patients agreed to measurement of oro-cecal transit time. Only three patients (15%) with abnormal lactose HBT might have had SIBO. The median oro-cecal transit time between LM and lactose intolerance patients were 75 min and 45 min, respectively (Z=2.545, P=0.011). CONCLUSIONS: Most of irritable bowel syndrome patients with an abnormal lactose HBT had LM. SIBO had little impact on the interpretation of lactose HBTs. The patients with lactose intolerance had faster small intestinal transit than LM patients.


Asunto(s)
Pruebas Respiratorias/métodos , Diarrea/diagnóstico , Diarrea/epidemiología , Intestino Delgado/microbiología , Síndrome del Colon Irritable/diagnóstico , Síndrome del Colon Irritable/epidemiología , Intolerancia a la Lactosa/diagnóstico , Adulto , Asia/epidemiología , Diarrea/microbiología , Diarrea/fisiopatología , Reacciones Falso Positivas , Femenino , Tránsito Gastrointestinal , Humanos , Hidrógeno , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/fisiopatología , Lactosa , Intolerancia a la Lactosa/epidemiología , Intolerancia a la Lactosa/microbiología , Intolerancia a la Lactosa/fisiopatología , Masculino , Persona de Mediana Edad , Prevalencia , Factores de Tiempo , Adulto Joven
14.
World J Gastroenterol ; 20(24): 7587-601, 2014 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-24976698

RESUMEN

Breath tests are non-invasive tests and can detect H2 and CH4 gases which are produced by bacterial fermentation of unabsorbed intestinal carbohydrate and are excreted in the breath. These tests are used in the diagnosis of carbohydrate malabsorption, small intestinal bacterial overgrowth, and for measuring the orocecal transit time. Malabsorption of carbohydrates is a key trigger of irritable bowel syndrome (IBS)-type symptoms such as diarrhea and/or constipation, bloating, excess flatulence, headaches and lack of energy. Abdominal bloating is a common nonspecific symptom which can negatively impact quality of life. It may reflect dietary imbalance, such as excess fiber intake, or may be a manifestation of IBS. However, bloating may also represent small intestinal bacterial overgrowth. Patients with persistent symptoms of abdominal bloating and distension despite dietary interventions should be referred for H2 breath testing to determine the presence or absence of bacterial overgrowth. If bacterial overgrowth is identified, patients are typically treated with antibiotics. Evaluation of IBS generally includes testing of other disorders that cause similar symptoms. Carbohydrate malabsorption (lactose, fructose, sorbitol) can cause abdominal fullness, bloating, nausea, abdominal pain, flatulence, and diarrhea, which are similar to the symptoms of IBS. However, it is unclear if these digestive disorders contribute to or cause the symptoms of IBS. Research studies show that a proper diagnosis and effective dietary intervention significantly reduces the severity and frequency of gastrointestinal symptoms in IBS. Thus, diagnosis of malabsorption of these carbohydrates in IBS using a breath test is very important to guide the clinician in the proper treatment of IBS patients.


Asunto(s)
Bacterias/metabolismo , Pruebas Respiratorias , Carbohidratos de la Dieta/metabolismo , Fermentación , Intestinos/microbiología , Síndrome del Colon Irritable/diagnóstico , Biomarcadores/metabolismo , Gases , Humanos , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/dietoterapia , Síndrome del Colon Irritable/microbiología , Intolerancia a la Lactosa/diagnóstico , Intolerancia a la Lactosa/microbiología , Valor Predictivo de las Pruebas , Pronóstico
15.
Nutr J ; 12: 160, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330605

RESUMEN

BACKGROUND: Lactose intolerance (LI) is a common medical problem with limited treatment options. The primary symptoms are abdominal pain, diarrhea, bloating, flatulence, and cramping. Limiting dairy foods to reduce symptoms contributes to low calcium intake and the risk for chronic disease. Adaptation of the colon bacteria to effectively metabolize lactose is a novel and potentially useful approach to improve lactose digestion and tolerance. RP-G28 is novel galacto-oligosaccharide (GOS) being investigated to improve lactose digestion and the symptoms of lactose intolerance in affected patients. METHODS: A randomized, double-blind, parallel group, placebo-controlled study was conducted at 2 sites in the United States. RP-G28 or placebo was administered to 85 patients with LI for 35 days. Post-treatment, subjects reintroduced dairy into their daily diets and were followed for 30 additional days to evaluate lactose digestion as measured by hydrogen production and symptom improvements via a patient-reported symptom assessment instrument. RESULTS: Lactose digestion and symptoms of LI trended toward improvement on RP-G28 at the end of treatment and 30 days post-treatment. A reduction in abdominal pain was also demonstrated in the study results. Fifty percent of RP-G28 subjects with abdominal pain at baseline reported no abdominal pain at the end of treatment and 30 days post treatment (p = 0.0190). RP-G28 subjects were also six times more likely to claim lactose tolerance post-treatment once dairy foods had been re-introduced into their diets (p = 0.0389). CONCLUSIONS: Efficacy trends and favorable safety/tolerability findings suggest that RP-G28 appears to be a potentially useful approach for improving lactose digestion and LI symptoms. The concurrent reduction in abdominal pain and improved overall tolerance could be a meaningful benefit to lactose intolerant individuals.


Asunto(s)
Digestión , Fármacos Gastrointestinales/uso terapéutico , Lactasa/deficiencia , Intolerancia a la Lactosa/dietoterapia , Lactosa/metabolismo , Oligosacáridos/uso terapéutico , Prebióticos , Dolor Abdominal/epidemiología , Dolor Abdominal/etiología , Dolor Abdominal/prevención & control , Adulto , Colon/microbiología , Productos Lácteos/efectos adversos , Método Doble Ciego , Femenino , Estudios de Seguimiento , Fármacos Gastrointestinales/administración & dosificación , Fármacos Gastrointestinales/efectos adversos , Humanos , Incidencia , Mucosa Intestinal/microbiología , Intolerancia a la Lactosa/microbiología , Intolerancia a la Lactosa/fisiopatología , Masculino , Oligosacáridos/administración & dosificación , Oligosacáridos/efectos adversos , Prebióticos/efectos adversos , Prebióticos/análisis , Índice de Severidad de la Enfermedad , Trisacáridos/administración & dosificación , Trisacáridos/efectos adversos , Trisacáridos/uso terapéutico , Estados Unidos/epidemiología
16.
Postepy Hig Med Dosw (Online) ; 67: 402-12, 2013 May 13.
Artículo en Polaco | MEDLINE | ID: mdl-23756375

RESUMEN

Beneficial effects due to the presence of probiotic bacteria of the genus Bifidobacterium in the human intestinal tract are still an interesting object of study. So far activities have been confirmed of bifidobacteria in stimulation of the host immune system, stimulation of tumor cell apoptosis, improvement of bowel motility, alleviation of symptoms of lactose intolerance, cholesterol lowering capacity, prevention and treatment of diarrhea and irritable bowel syndrome, alleviation of allergy or atopic dermatitis, maintenance of homeostasis of the intestine, and stimulation of the development of normal intestinal microflora in infants. A multitude of therapeutic properties encourages researchers to investigate the possibility of using the potential of Bifidobacterium in the prevention and treatment of other conditions such as rheumatoid arthritis and depression. Although it is known that the beneficial effects are due to intestinal mucosal colonization by these bacteria, the cell components responsible for the colonization are still not determined. In addition to the beneficial effects of probiotic administration, there were also negative effects including sepsis. Therefore research has been directed to identify specific components of Bifidobacterium responsible for probiotic effects. Currently researchers are focused on identifying, isolating and evaluating the properties of surface proteins that are probably involved in the adhesion of bacterial cells to the intestinal epithelium, improving colonization. This paper is an overview of current knowledge on Bifidobacterium surface proteins. The ways of transport and anchoring proteins in Gram-positive bacterial cells, the assembly of cell wall, and a description of the genus Bifidobacterium are presented.


Asunto(s)
Bifidobacterium/metabolismo , Mucosa Intestinal/microbiología , Proteínas de la Membrana/metabolismo , Bifidobacterium/clasificación , Bifidobacterium/crecimiento & desarrollo , Diarrea/microbiología , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/microbiología , Inmunización , Lactante , Mucosa Intestinal/inmunología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/prevención & control , Lactobacillus/crecimiento & desarrollo , Lactobacillus/metabolismo , Intolerancia a la Lactosa/microbiología , Intolerancia a la Lactosa/prevención & control , Especificidad de la Especie
17.
Eur Rev Med Pharmacol Sci ; 17 Suppl 2: 18-25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24443063

RESUMEN

This review discusses one of the most relevant problems in gastrointestinal clinical practice: lactose intolerance. The role of lactase-persistence alleles the diagnosis of lactose malabsorption the development of lactose intolerance symptoms and its management. Most people are born with the ability to digest lactose, the major carbohydrate in milk and the main source of nutrition until weaning. Approximately, 75% of the world's population loses this ability at some point, while others can digest lactose into adulthood. Symptoms of lactose intolerance include abdominal pain, bloating, flatulence and diarrhea with a considerable intraindividual and interindividual variability in the severity. Diagnosis is most commonly performed by the non invasive lactose hydrogen breath test. Management of lactose intolerance consists of two possible clinical choice not mutually exclusive: alimentary restriction and drug therapy.


Asunto(s)
Pruebas Respiratorias , Dieta Baja en Carbohidratos , Terapia de Reemplazo Enzimático , Lactasa/uso terapéutico , Intolerancia a la Lactosa/diagnóstico , Intolerancia a la Lactosa/terapia , Lactosa/metabolismo , Bacterias/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Hidrólisis , Intestinos/microbiología , Lactasa/genética , Lactasa/metabolismo , Intolerancia a la Lactosa/enzimología , Intolerancia a la Lactosa/genética , Intolerancia a la Lactosa/microbiología , Fenotipo , Valor Predictivo de las Pruebas , Resultado del Tratamiento
18.
Appl Microbiol Biotechnol ; 96(6): 1499-506, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22395907

RESUMEN

The endogenous ß-galactosidase expressed in intestinal microbes is demonstrated to help humans in lactose usage, and treatment associated with the promotion of beneficial microorganism in the gut is correlated with lactose tolerance. From this point, a kind of recombinant live ß-galactosidase delivery system using food-grade protein expression techniques and selected probiotics as vehicle was promoted by us for the purpose of application in lactose intolerance subjects. Previously, a recombinant Lactococcus lactis MG1363 strain expressing food-grade ß-galactosidase, the L. lactis MG1363/FGZW, was successfully constructed and evaluated in vitro. This study was conducted to in vivo evaluate its efficacy on alleviating lactose intolerance symptoms in post-weaning Balb/c mice, which were orally administered with 1 × 106 CFU or 1 × 108 CFU of L. lactis MG1363/FGZW daily for 4 weeks before lactose challenge. In comparison with naïve mice, the mice administered with L. lactis MG1363/FGZW showed significant alleviation of diarrhea symptoms in less total feces weight within 6 h post-challenge and suppressed intestinal motility after lactose challenge, although there was no significant increase of ß-galactosidase activity in small intestine. The alleviation also correlated with higher species abundance, more Bifidobacterium colonization, and stronger colonization resistance in mice intestinal microflora. Therefore, this recombinant L. lactis strain effectively alleviated diarrhea symptom induced by lactose uptake in lactose intolerance model mice with the probable mechanism of promotion of lactic acid bacteria to differentiate and predominantly colonize in gut microbial community, thus making it a promising probiotic for lactose intolerance subjects.


Asunto(s)
Terapia Biológica , Lactococcus lactis/genética , Intolerancia a la Lactosa/terapia , Probióticos/administración & dosificación , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo , Animales , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Expresión Génica , Humanos , Lactococcus lactis/metabolismo , Lactosa/metabolismo , Intolerancia a la Lactosa/metabolismo , Intolerancia a la Lactosa/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Destete
19.
Acta Gastroenterol Latinoam ; 40(3): 221-4, 2010 Sep.
Artículo en Español | MEDLINE | ID: mdl-21053480

RESUMEN

INTRODUCTION: Some patients complain of digestive symptoms related to diary products intake. This intolerance could be associated with an intestinal bacterial overgrowth or an increased fermentative intestinal profile and not due to lactose malabsorption. OBJECTIVE: To estimate the prevalence of bacterial overgrowth in subjects with digestive symptoms related to diary products intolerance. MATERIAL AND METHODS: Patients who had performed hydrogen breath test due to chronic functional distension syndrome (Rome III) were analyzed. Thirty of them (22 female, average age 52 years) complained of symptoms related to diary products intake. All subjects completed a nutritional survey that allowed to assess the degree of tolerance to milk products and performed the hydrogen breath test with lactulose as substrate. Bacterial overgrowth was considered when baseline values were over 15 parts per million (ppm), values before 80 minutes were greater than 20 ppm or values of area under the curve were greater than 3,000 ppm/min in the 180 studied minutes. RESULTS: Fifty four patients were analyzed. Thirty of them showed moderate, important or severe clinical milk intolerance. Of these patients, 23 (77%), had a positive breath hydrogen test according to used criteria. CONCLUSIONS: The prevalence of small bowel bacterial overgrowth or an increased fermentative intestinal profile among individuals who complain of symptoms related to diary products is high and this fact should be considered in order to avoid empirical restrictive diets.


Asunto(s)
Bacterias/crecimiento & desarrollo , Intestino Delgado/microbiología , Intolerancia a la Lactosa/microbiología , Adulto , Anciano , Pruebas Respiratorias , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Estudios Prospectivos , Índice de Severidad de la Enfermedad
20.
Toxicology ; 278(3): 268-76, 2010 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-20851732

RESUMEN

Lactose and food intolerance cause a wide range of gut and systemic symptoms, including gas, gut pain, diarrhoea or constipation, severe headaches, severe fatigue, loss of cognitive functions such as concentration, memory and reasoning, muscle and joint pain, heart palpitations, and a variety of allergies (Matthews and Campbell, 2000; Matthews et al., 2005; Waud et al., 2008). These can be explained by the production of toxic metabolites from gut bacteria, as a result of anaerobic digestion of carbohydrates and other foods, not absorbed in the small intestine. These metabolites include alcohols, diols such as butan 2,3 diol, ketones, acids, and aldehydes such as methylglyoxal (Campbell et al., 2005, 2009). These 'toxins' induce calcium signals in bacteria and affect their growth, thereby acting to modify the balance of microflora in the gut (Campbell et al., 2004, 2007a,b). These bacterial 'toxins' also affect signalling mechanisms in cells around the body, thereby explaining the wide range of symptoms in people with food intolerance. This new mechanism also explains the most common referral to gastroenterologists, irritable bowel syndrome (IBS), and the illness that afflicted Charles Darwin for 50 years (Campbell and Matthews, 2005a,b). We propose it will lead to a new understanding of the molecular mechanism of type 2 diabetes and some cancers.


Asunto(s)
Bacterias/metabolismo , Carbohidratos de la Dieta/toxicidad , Alimentos , Enfermedades Gastrointestinales/microbiología , Síndrome del Colon Irritable/microbiología , Intolerancia a la Lactosa/microbiología , Bacterias/efectos de los fármacos , Toxinas Bacterianas/toxicidad , Señalización del Calcio/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Carbohidratos de la Dieta/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Piruvaldehído/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...