Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
J Nutr Biochem ; 99: 108865, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34582967

RESUMEN

Pain-related functional gastrointestinal disorders (FGIDs) are characterized by visceral hypersensitivity (VHS) associated with alterations in the microbiota-gut-brain axis. Since human milk oligosaccharides (HMOs) modulate microbiota, gut and brain, we investigated whether HMOs impact VHS, and explored the role of gut microbiota. To induce VHS, C57BL/6JRj mice received hourly water avoidance stress (WAS) sessions for 10 d, or antibiotics (ATB) for 12 d. Challenged and unchallenged (Sham) animals were fed AIN93M diet (Cont) or AIN93M containing 1% of a 6-HMO mix (HMO6). VHS was assessed by monitoring the visceromotor response to colorectal distension. Fecal microbiome was analyzed by shotgun metagenomics. The effect of HMO6 sub-blends on VHS and nociceptive pathways was further tested using the WAS model. In mice fed Cont, WAS and ATB increased the visceromotor response to distension. HMO6 decreased WAS-mediated electromyographic rise at most distension volumes and overall Area Under Curve (AUC=6.12±0.50 in WAS/HMO6 vs. 9.46±0.50 in WAS/Cont; P<.0001). In contrast, VHS in ATB animals was not improved by HMO6. In WAS, HMO6 promoted most microbiota taxa and several functional pathways associated with low VHS and decreased those associated with high VHS. Among the sub-blends, 2'FL+DFL and LNT+6'SL reduced visceromotor response close to Sham/Cont values and modulated serotoninergic and CGRPα-related pathways. This research further substantiates the capacity of HMOs to modulate the microbiota-gut-brain communication and identifies mitigation of abdominal pain as a new HMO benefit. Ultimately, our findings suggest the value of specific HMO blends to alleviate pain associated FGIDs such as infantile colic or Irritable Bowel Syndrome.


Asunto(s)
Dolor Abdominal/dietoterapia , Disbiosis/dietoterapia , Microbioma Gastrointestinal , Leche Humana/metabolismo , Oligosacáridos/metabolismo , Dolor Abdominal/metabolismo , Dolor Abdominal/microbiología , Dolor Abdominal/psicología , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/psicología , Heces/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Oligosacáridos/análisis , Estrés Psicológico
2.
J Immunol Res ; 2021: 5578958, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34869782

RESUMEN

BACKGROUND: As a transitional state between normal aging and Alzheimer's disease (AD), mild cognitive impairment (MCI) is characterized by a worse cognitive decline than that of natural aging. The association between AD and gut microbiota has been reported in a number of studies; however, microbial research regarding MCI remains limited. METHODS: This study examined 48 participants, of whom 22 were MCI cases and 26 were normal control cases. Fecal samples were collected for 16S ribosomal RNA (rRNA) quantitative arrays and bioinformatics analysis. RESULTS: A principal coordinates analysis (PCoA) and nonmetric multidimensional scaling (NMDS) both demonstrated that the microbial composition of participants with MCI deviated from that of healthy control participants. Multiple bacterial species were significantly increased (e.g., Staphylococcus intermedius) or decreased (e.g., Bacteroides salyersiae) in samples from the MCI group. CONCLUSION: The composition of gut microbiota differed between normal control and MCI cases. This is the first study to identify a signature series of species in the gut microbiota of individuals with MCI. The results provide a new direction for the future development of an early diagnosis and probiotic regimen.


Asunto(s)
Envejecimiento/inmunología , Disfunción Cognitiva/inmunología , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Anciano , Estudios de Casos y Controles , Disfunción Cognitiva/microbiología , Disfunción Cognitiva/prevención & control , Disbiosis/dietoterapia , Disbiosis/inmunología , Disbiosis/microbiología , Heces/microbiología , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Probióticos/administración & dosificación
3.
Biomolecules ; 11(8)2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34439821

RESUMEN

Irritable bowel syndrome is not a life-threatening disease, yet it significantly affects the quality of life and contributes to economic loss. It is estimated that even up to 45% of the world's population can suffer from the disease. The first attempts to diagnose irritable bowel syndrome were made at the end of the 19th century; however, establishing appropriate diagnostic criteria and treatment methods is still ongoing. To date, little is known about the etiology of irritable bowel syndrome; however, growing attention is drawn to the intestinal microbiota as a factor in the disease development. For this reason, researchers have conducted many studies on therapies that modulate the microbiota, among which probiotics, prebiotics, and synbiotics are widely studied. To date, most studies have examined probiotics; however, there are also several studies demonstrating the efficacy of prebiotics and synbiotics. The aim of this review was to summarize findings on the usefulness of probiotics, prebiotics, and synbiotics in the treatment of irritable bowel syndrome.


Asunto(s)
Disbiosis/dietoterapia , Microbioma Gastrointestinal/efectos de los fármacos , Síndrome del Colon Irritable/dietoterapia , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Simbióticos/administración & dosificación , Actinobacteria/genética , Actinobacteria/crecimiento & desarrollo , Actinobacteria/aislamiento & purificación , Antibacterianos/efectos adversos , Bacteroidetes/genética , Bacteroidetes/crecimiento & desarrollo , Bacteroidetes/aislamiento & purificación , Ensayos Clínicos como Asunto , Disbiosis/etiología , Disbiosis/microbiología , Disbiosis/patología , Firmicutes/genética , Firmicutes/crecimiento & desarrollo , Firmicutes/aislamiento & purificación , Fusobacterias/genética , Fusobacterias/crecimiento & desarrollo , Fusobacterias/aislamiento & purificación , Microbioma Gastrointestinal/genética , Humanos , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/patología , Proteobacteria/genética , Proteobacteria/crecimiento & desarrollo , Proteobacteria/aislamiento & purificación , Calidad de Vida
4.
Reprod Biomed Online ; 43(3): 503-514, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34315695

RESUMEN

RESEARCH QUESTION: Does oral probiotic supplementation influence the relative abundance of different vaginal microbiota in women experiencing infertility? DESIGN: A prospective, monocentric randomized controlled trial. To study the influence of probiotics on infertility, 80 patients with primary or secondary infertility were included. Patients were assigned to either a probiotic treatment or a control group. Participants in the treatment group (n = 40) took one sachet (2 g) a day of a defined probiotic supplement limiting Lactobacillus strains. Patients in the control group did not receive any additional probiotic supplements. Vaginal samples were taken on day 20 of the menstrual cycle and 4 weeks later, on day 20, of the consecutive cycle. Subsequently, 16s rRNA gene analysis of the vaginal samples was conducted. RESULTS: After the intervention phase, no effects on alpha diversity resulting from treatment could be observed. The between sample diversity of different women (beta diversity) at baseline had no effects of age, treatment group or body mass index. Primary or secondary sterility, however, had a significant effect on community. Three clusters (Lactobacillus crispatus, Lactobacillus iners and Lactobacillus gasseri) were identified as the leading representatives. Furthermore, patients treated with probiotics showed limited growth of Ureaplasma parvum compared with the control group (P = 0.021). CONCLUSIONS: This study points to a possible protective effect of probiotic supplements on the vaginal microbiota. It is tempting to speculate that this effect assists in containing the growth of non-beneficial bacteria and helps to prevent or cure a dysbiotic vaginal flora.


Asunto(s)
Infertilidad Femenina/dietoterapia , Probióticos/farmacología , Infecciones por Ureaplasma/dietoterapia , Vagina/efectos de los fármacos , Vaginosis Bacteriana/dietoterapia , Adolescente , Adulto , Austria , Suplementos Dietéticos , Disbiosis/complicaciones , Disbiosis/dietoterapia , Femenino , Humanos , Infertilidad Femenina/microbiología , Lactobacillus/fisiología , Probióticos/administración & dosificación , Ureaplasma/efectos de los fármacos , Infecciones por Ureaplasma/complicaciones , Vagina/microbiología , Vaginosis Bacteriana/complicaciones , Adulto Joven
5.
Nutrients ; 13(7)2021 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-34202894

RESUMEN

Consumption of different types of high-calorie foods leads to the development of various metabolic disorders. However, the effects of multi-strain probiotics on different types of diet-induced obesity and intestinal dysbiosis remain unclear. In this study, mice were fed a control diet, high-fat diet (HFD; 60% kcal fat and 20% kcal carbohydrate), or western diet (WD; 40% kcal fat and 43% kcal carbohydrate) and administered with multi-strain AB-Kefir containing six strains of lactic acid bacteria and a Bifidobacterium strain, at 109 CFU per mouse for 10 weeks. Results demonstrated that AB-Kefir reduced body weight gain, glucose intolerance, and hepatic steatosis with a minor influence on gut microbiota composition in HFD-fed mice, but not in WD-fed mice. In addition, AB-Kefir significantly reduced the weight and size of adipose tissues by regulating the expression of CD36, Igf1, and Pgc1 in HFD-fed mice. Although AB-Kefir did not reduce the volume of white adipose tissue, it markedly regulated CD36, Dgat1 and Mogat1 mRNA expression. Moreover, the abundance of Eubacterium_coprostanoligenes_group and Ruminiclostridium significantly correlated with changes in body weight, liver weight, and fasting glucose in test mice. Overall, this study provides important evidence to understand the interactions between probiotics, gut microbiota, and diet in obesity treatment.


Asunto(s)
Dieta de Carga de Carbohidratos/métodos , Dieta Alta en Grasa/métodos , Kéfir/microbiología , Obesidad/dietoterapia , Probióticos/administración & dosificación , Tejido Adiposo/efectos de los fármacos , Animales , Peso Corporal , Dieta de Carga de Carbohidratos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Dieta Occidental , Sacarosa en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Disbiosis/dietoterapia , Disbiosis/microbiología , Hígado Graso/dietoterapia , Hígado Graso/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Intolerancia a la Glucosa/dietoterapia , Intolerancia a la Glucosa/microbiología , Inflamación , Hígado/patología , Ratones , Ratones Obesos , Obesidad/etiología , Obesidad/microbiología , Aumento de Peso/efectos de los fármacos
6.
Behav Brain Res ; 414: 113474, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34280457

RESUMEN

Research has established that stress "gets under the skin," impacting neuroendocrine and neuroimmune pathways to influence risk for physical and mental health outcomes. These effects can be particularly significant for early life stress (ELS), or adverse childhood experiences (ACEs). In this review, we explore whether stress gets "into the belly," that is, whether psychosocial stress affects the gut microbiome. We review animal and human research utilizing a variety of stress paradigms (acute laboratory stressors, chronic stress, stressful life events, perceived stress, ELS, in utero stress) and their impacts on the gut microbiota, with a particular focus on ELS. We also review data on dietary interventions to moderate impact of stress on the gut microbiome. Our review suggests strong evidence that acute laboratory stress, chronic stress, and ELS affect the gut microbiota in rodents, and growing evidence that perceived stress and ELS may impact the gut microbiota in humans. Emerging data also suggests, particularly in rodents, that dietary interventions such as omega-3 fatty acids and pre- and pro-biotics may buffer against the effects of stress on the gut microbiome, but more research is needed. In sum, growing evidence suggests that stress impacts not only the neuroendocrine and neuroimmune axes, but also the microbiota-gut-brain-axis, providing a pathway by which stress may get "into the belly" to influence health risk.


Asunto(s)
Experiencias Adversas de la Infancia , Disbiosis , Microbioma Gastrointestinal , Estrés Psicológico , Animales , Disbiosis/dietoterapia , Disbiosis/etiología , Disbiosis/microbiología , Humanos , Estrés Psicológico/complicaciones , Estrés Psicológico/microbiología
7.
Nutrients ; 13(5)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946961

RESUMEN

Irritable Bowel Syndrome (IBS) is a chronic functional gastrointestinal disorder characterized by abdominal pain associated with defecation or a change in bowel habits. Gut microbiota, which acts as a real organ with well-defined functions, is in a mutualistic relationship with the host, harvesting additional energy and nutrients from the diet and protecting the host from pathogens; specific alterations in its composition seem to play a crucial role in IBS pathophysiology. It is well known that diet can significantly modulate the intestinal microbiota profile but it is less known how different nutritional approach effective in IBS patients, such as the low-FODMAP diet, could be responsible of intestinal microbiota changes, thus influencing the presence of gastrointestinal (GI) symptoms. The aim of this review was to explore the effects of different nutritional protocols (e.g., traditional nutritional advice, low-FODMAP diet, gluten-free diet, etc.) on IBS-D symptoms and on intestinal microbiota variations in both IBS-D patients and healthy subjects. To date, an ideal nutritional protocol does not exist for IBS-D patients but it seems crucial to consider the effect of the different nutritional approaches on the intestinal microbiota composition to better define an efficient strategy to manage this functional disorder.


Asunto(s)
Diarrea/dietoterapia , Diarrea/etiología , Disbiosis/dietoterapia , Síndrome del Colon Irritable/clasificación , Síndrome del Colon Irritable/dietoterapia , Dieta , Microbioma Gastrointestinal , Humanos , Síndrome del Colon Irritable/patología
8.
Food Chem Toxicol ; 153: 112306, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34058235

RESUMEN

Nowadays, people are exposed to diverse environmental and chemical pollutants produced by industry and agriculture. Food contaminations such as persistent organic pollutants (POPs), heavy metals, and mycotoxins are a serious concern for global food safety with economic and public health implications especially in the newly industrialized countries (NIC). Mounting evidence indicates that chronic exposure to food contaminants referred to as xenobiotics exert a negative effect on human health such as inflammation, oxidative stress, and intestinal disorders linked with perturbation of the composition and metabolic profile of the gut microflora. Although the physicochemical technologies for food decontamination are utilized in many cases but require adequate conditions which are often not feasible to be met in many industrial sectors. At present, one promising approach to reduce the risk related to the presence of xenobiotics in foodstuffs is a biological detoxification done by probiotic strains and their enzymes. Many studies confirmed that probiotics are an effective, feasible, and inexpensive tool for preventing xenobiotic-induced dysbiosis and alleviating their toxicity. This review aims to summarize the current knowledge of the direct mechanisms by which probiotics can influence the detoxification of xenobiotics. Moreover, probiotic-xenobiotic interactions with the gut microbiota and the host response were also discussed.


Asunto(s)
Contaminación de Alimentos/prevención & control , Probióticos/metabolismo , Xenobióticos/metabolismo , Animales , Disbiosis/inducido químicamente , Disbiosis/dietoterapia , Microbioma Gastrointestinal/fisiología , Expresión Génica/fisiología , Humanos , Probióticos/uso terapéutico , Xenobióticos/toxicidad
9.
PLoS One ; 16(4): e0250423, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33914799

RESUMEN

The tight association between malnutrition and gut microbiota (GM) dysbiosis enables microbiota-targeting intervention to be a promising strategy. Thus, we used a malnourished pig model to investigate the host response and GM alterations under different diet supplementation strategies. Pigs at age of 4 weeks were fed with pure maize diet to induce malnutrition symptoms, and followed by continuous feeding with maize (Maize, n = 8) or re-feeding using either corn-soy-blend (CSB+, n = 10) or millet-soy-blend based (MSB+, n = 10) supplementary food for 3 weeks. Meanwhile, 8 pigs were fed on a standard formulated ration as control (Ref). The effect of nutritional supplementation was assessed by the growth status, blood chemistry, gastrointestinal pathology, mucosal microbiota composition and colon production of short-chain fatty acids. Compared with purely maize-fed pigs, both CSB+ and MSB+ elevated the concentrations of total protein and globulin in blood. These pigs still showed most malnutrition symptoms after the food intervention period. MSB+ had superior influence on the GM development, exhibiting better performance in both structural and functional aspects. MSB+ pigs were colonized by less Proteobacteria but more Bacteroidetes, Firmicutes and Lachnospira spp. Pearson's correlation analysis indicated a strong correlation between the abundance of mucosal e.g., Faecalibacterium and Lachnospira spp. and body weight, crown-rump length and total serum protein. In conclusion, the malnutrition symptoms were accompanied by an aberrant GM, and millet-based nutritional supplementation showed promising potentials to restore the reduced GM diversity implicated in pig malnutrition.


Asunto(s)
Alimentación Animal/análisis , Dieta/métodos , Disbiosis/dietoterapia , Microbioma Gastrointestinal/fisiología , Desnutrición/dietoterapia , Mijos/química , Animales , Bacteroidetes/genética , Bacteroidetes/crecimiento & desarrollo , Bacteroidetes/aislamiento & purificación , Biodiversidad , Proteínas Sanguíneas/agonistas , Proteínas Sanguíneas/metabolismo , Peso Corporal , Clostridiales/genética , Clostridiales/crecimiento & desarrollo , Clostridiales/aislamiento & purificación , Disbiosis/microbiología , Disbiosis/patología , Faecalibacterium/genética , Faecalibacterium/crecimiento & desarrollo , Faecalibacterium/aislamiento & purificación , Ácidos Grasos Volátiles/biosíntesis , Femenino , Firmicutes/genética , Firmicutes/crecimiento & desarrollo , Firmicutes/aislamiento & purificación , Desnutrición/microbiología , Desnutrición/patología , Proteobacteria/genética , Proteobacteria/crecimiento & desarrollo , Proteobacteria/aislamiento & purificación , ARN Ribosómico 16S/genética , Glycine max/química , Porcinos , Verrucomicrobia/genética , Verrucomicrobia/crecimiento & desarrollo , Verrucomicrobia/aislamiento & purificación , Zea mays/química
10.
Sci Rep ; 11(1): 6367, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737543

RESUMEN

Obesity, a major healthcare problem worldwide, induces metabolic endotoxemia through the gut translocation of lipopolysaccharides (LPS), a major cell wall component of Gram-negative bacteria, causing a chronic inflammatory state. A combination of several probiotics including Lactobacillus acidophilus 5 (LA5), a potent lactic acid-producing bacterium, has previously been shown to attenuate obesity. However, data on the correlation between a single administration of LA5 versus microbiota alteration might be helpful for the probiotic adjustment. LA5 was administered daily together with a high-fat diet (HFD) for 8 weeks in mice. Furthermore, the condition media of LA5 was also tested in a hepatocyte cell-line (HepG2 cells). Accordingly, LA5 attenuated obesity in mice as demonstrated by weight reduction, regional fat accumulation, lipidemia, liver injury (liver weight, lipid compositions, and liver enzyme), gut permeability defect, endotoxemia, and serum cytokines. Unsurprisingly, LA5 improved these parameters and acidified fecal pH leads to the attenuation of fecal dysbiosis. The fecal microbiome analysis in obese mice with or without LA5 indicated; (i) decreased Bacteroidetes (Gram-negative anaerobes that predominate in non-healthy conditions), (ii) reduced total fecal Gram-negative bacterial burdens (the sources of gut LPS), (iii) enhanced Firmicutes (Gram-positive bacteria with potential benefits) and (iv) increased Verrucomycobia, especially Akkermansia muciniphila, a bacterium with the anti-obesity property. With LA5 administration, A. muciniphila in the colon were more than 2,000 folds higher than the regular diet mice as determined by 16S rRNA. Besides, LA5 produced anti-inflammatory molecules with a similar molecular weight to LPS that reduced cytokine production in LPS-activated HepG2 cells. In conclusion, LA5 attenuated obesity through (i) gut dysbiosis attenuation, partly through the promotion of A. muciniphila (probiotics with the difficulty in preparation processes), (ii) reduced endotoxemia, and (iii) possibly decreased liver injury by producing the anti-inflammatory molecules.


Asunto(s)
Disbiosis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/tratamiento farmacológico , Probióticos/farmacología , Akkermansia/efectos de los fármacos , Akkermansia/crecimiento & desarrollo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/dietoterapia , Disbiosis/microbiología , Disbiosis/patología , Humanos , Lactobacillus acidophilus/química , Lactobacillus acidophilus/metabolismo , Ratones , Ratones Obesos , Obesidad/etiología , Obesidad/microbiología , Obesidad/patología , Probióticos/química , ARN Ribosómico 16S/genética
11.
Medicine (Baltimore) ; 100(10): e18621, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33725807

RESUMEN

INTRODUCTION: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with increasing incidence. The externalizing and internalizing problems among children with ASD often persistent and highly impair functioning of both the child and the family. Children with ASD often develop gut-related comorbidities and dysbiosis can have negative effects on not only the gastrointestinal (GI) tract, but also psychological symptoms. Dietary exclusions and probiotic supplements also have been investigated in the management of ASD symptoms. Especially, there is some anecdotal evidence that probiotics supplements are able to alleviate GI symptoms as well as improve behaviors in children with ASD. METHOD AND ANALYSIS: This review will report on overall studies that include randomized control trials, randomized cross-over studies and cluster-randomized trials designs that consider curative effect in children with ASD by probiotic supplements. We will search 6 databases: MEDLINE, Embase, Scopus, PubMed, The Cochrane Library, and Web of Science and we will perform a manual search the journal Autism and information of ongoing or unpublished studies. The Mixed Methods Appraisal Tool (MMAT) will be used to assess quality of articles and the Jadad scale will be used to assess for bias. Assessment of publication bias will be performed using funnel plots generated by Comprehensive Meta-Analysis (CMA) 3.0 software. Clarifying the evidence in this area will be important for future research directions when reformulating and promoting the therapeutic regime in the field. ETHICS AND DISSEMINATION: There are no human participants, data, or tissue being directly studied for the purposes of the review; therefore, ethics approval and consent to participate are not applicable. The results of this study will be presented at conferences and published in peer-reviewed journals. REGISTRATION AND STATUS: PROSPERO 2019 CRD42019132754.


Asunto(s)
Trastorno del Espectro Autista/dietoterapia , Disbiosis/dietoterapia , Probióticos/administración & dosificación , Trastorno del Espectro Autista/epidemiología , Trastorno del Espectro Autista/psicología , Niño , Comorbilidad , Disbiosis/epidemiología , Disbiosis/psicología , Humanos , Metaanálisis como Asunto , Revisiones Sistemáticas como Asunto , Resultado del Tratamiento
12.
Food Funct ; 12(7): 3104-3119, 2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33725036

RESUMEN

Antibiotics are effective treatments for bacterial infections, however, their oral administration can have unintended consequences and may alter the gut microbiota composition. In this study, we examined the influence of antibiotics on the induction of gut dysbiosis and then evaluated the potential of cow and goat milk to restore the microbiota composition and metabolism in newly weaned rats. In the first study (gut dysbiosis model), rats were treated with amoxicillin, a mixture of antibiotics (ampicillin, gentamicin and metronidazole) or no antibiotics (control). Antibiotics reduced the rat body weights, food intakes and faecal outputs compared to the control group. Gut length was significantly decreased after the antibiotic intake. The bacterial populations (Bifidobacterium spp., Lactobacillus spp. and total bacteria) and short-chain fatty acids (SCFAs; acetic, butyric and propionic) concentrations in rat caecum, colon and faeces were significantly altered by the antibiotic treatments. In the second study, we examined the effects of cow and goat milk in restoring bacterial populations and metabolism in rats with gut dysbiosis induced by amoxicillin. Goat milk significantly increased the numbers of Bifidobacterium spp. and Lactobacillus spp. and decreased the numbers of Clostridium perfringens in the caecum and colon of rats treated with amoxicillin. Whereas, rats fed cow milk had higher Lactobacillus spp. and lower C. perfringens in the gut. Caecal and colonic SCFAs (acetic, butyric and propionic) concentrations differed significantly between rats fed cow and goat milk diets. Overall, goat and cow milk varied in their effects on the immature gut following antibiotic-induced dysbiosis in a rat model.


Asunto(s)
Disbiosis/dietoterapia , Leche/microbiología , Amoxicilina , Animales , Bovinos , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Fermentación , Microbioma Gastrointestinal/efectos de los fármacos , Cabras , Masculino , Ratas , Ratas Sprague-Dawley
13.
Mar Drugs ; 19(2)2021 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-33498729

RESUMEN

Several cardioprotective mechanisms attributed to Omega-3 polyunsaturated fatty acids (PUFAs) have been studied and widely documented. However, in recent years, studies have supported the concept that the intestinal microbiota can play a much larger role than we had anticipated. Microbiota could contribute to several pathologies, including cardiovascular diseases. Indeed, an imbalance in the microbiota has often been reported in patients with cardiovascular disease and produces low-level inflammation. This inflammation contributes to, more or less, long-term development of cardiovascular diseases. It can also worsen the symptoms and the consequences of these pathologies. According to some studies, omega-3 PUFAs in the diet could restore this imbalance and mitigate its harmful effects on cardiovascular diseases. Many mechanisms are involved and included: (1) a reduction of bacteria producing trimethylamine (TMA); (2) an increase in bacteria producing butyrate, which has anti-inflammatory properties; and (3) a decrease in the production of pro-inflammatory cytokines. Additionally, omega-3 PUFAs would help maintain better integrity in the intestinal barrier, thereby preventing the translocation of intestinal contents into circulation. This review will summarize the effects of omega-3 PUFAs on gut micro-biota and the potential impact on cardiac health.


Asunto(s)
Cardiotónicos/administración & dosificación , Enfermedades Cardiovasculares/dietoterapia , Ácidos Grasos Omega-3/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Cardiotónicos/metabolismo , Enfermedades Cardiovasculares/metabolismo , Dieta Saludable/métodos , Dieta Saludable/tendencias , Disbiosis/dietoterapia , Disbiosis/metabolismo , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/administración & dosificación , Ácidos Grasos Omega-6/efectos adversos , Microbioma Gastrointestinal/fisiología , Humanos
14.
Artículo en Inglés | MEDLINE | ID: mdl-32949638

RESUMEN

Alzheimer's disease (AD) is a complex multifactorial disease involving chronic neuroinflammation and neurodegeneration. It has been recently recognized that gut microbiota interacts with the brain, and it is termed as microbiota-gut-brain axis. Modulation of this axis has been recently reported to affect the pathogenesis of neurodegenerative diseases, such as AD. Gut microbiota has a pivotal role in regulating multiple neuro-chemical pathways through the highly interconnected gut-brain axis. Recent emerging evidences have highlighted that the intestinal microflora takes part in bidirectional communication between the gut and the brain. Due to this, the researchers have suggested that human gut microflora may even act as the "second brain" and may be responsible for neurodegenerative disorders like Alzheimer's disease. Dysbiosis of gut microbiota can induce increased intestinal permeability and systemic inflammation. This may lead to the development of AD pathologies and cognitive impairment via the neural, immune, endocrine, and metabolic pathways. Thus, the modulation of gut microbiota through personalized diet, oral bacteriotherapy may lead to alteration of gut microbiota their products including amyloid protein. It has been demonstrated that modulation of the gut microbiota induces beneficial effects on neuronal pathways consequently leading to delay the progression of Alzheimer's disease. Thus, this approach may provide a novel therapeutic option for treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Eje Cerebro-Intestino/fisiología , Encéfalo/metabolismo , Disbiosis/metabolismo , Microbioma Gastrointestinal/fisiología , Mediadores de Inflamación/metabolismo , Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/patología , Animales , Disbiosis/dietoterapia , Disbiosis/patología , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Enfermedades Neuroinflamatorias/dietoterapia , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Probióticos/administración & dosificación
15.
Artículo en Inglés | MEDLINE | ID: mdl-32898589

RESUMEN

As a neuropsychiatric disorder, substance addiction represents a major public health issue with high prevalence and mortality in many countries. Recently, gut microbiota has been certified to play a part in substance addiction through various mechanisms. Hence, we mainly focused on three substance including alcohol, cocaine and methamphetamine in this review, and summarized their relationships with gut microbiota, respectively. Besides, we also concluded the possible treatments for substance addiction from the perspective of applying gut microbiota. This review aims to build a bridge between substance addiction and gut microbiota according to existing evidences, so as to excavate the possible bi-directional function of microbiota-gut-brain axis in substance addiction for developing therapeutic strategies in the future.


Asunto(s)
Eje Cerebro-Intestino/fisiología , Disbiosis/metabolismo , Microbioma Gastrointestinal/fisiología , Trastornos Relacionados con Sustancias/metabolismo , Alcoholismo/complicaciones , Alcoholismo/metabolismo , Eje Cerebro-Intestino/efectos de los fármacos , Cocaína/efectos adversos , Disbiosis/complicaciones , Disbiosis/dietoterapia , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Metanfetamina/efectos adversos , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Trastornos Relacionados con Sustancias/complicaciones , Trastornos Relacionados con Sustancias/dietoterapia
16.
Antioxid Redox Signal ; 34(10): 811-830, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32316741

RESUMEN

Significance: Hypertension (HTN) has a complex etiology that is characterized by genetic and environmental factors. It has become a global health burden leading to cardiovascular diseases and kidney diseases, ultimately progressing to premature death. Accumulating evidence indicated that gut microbiome was associated with metabolic disorders and inflammation, which were closely linked to HTN. Recent Advances: Recent studies using bacterial genomic analysis and fecal microbiota transplantation as well as many lines of seminal evidence demonstrated that aberrant gut microbiome was significantly associated with HTN. The intestinal microbiome of both patients and animals with HTN had decreased bacterial diversity, disordered microbial structure and functions, and altered end products of fermentation. Gut dysbiosis and metabolites of the gut microbiota play an important role in blood pressure (BP) control, and they are therefore responsible for developing HTN. Critical Issues: This study aimed at focusing on the recent advances in understanding the role played by gut bacteria and the mechanisms underlying the pathological milieu that induced elevated BP and led to HTN pathogenesis. Potential intervention strategies targeting the correction of gut dysbiosis to improve HTN development were summarized. Future Directions: Larger numbers of fecal transplants from participants with HTN should be carried out to examine the magnitude of BP changes with the replacement of the gut microbiome. The proposed mechanisms for the gut in regulating BP remain to be verified. Whether intervention strategies using probiotics, dietary interventions, bacteriophages, and fecal transplants are feasible for individuals with HTN remains to be explored. Antioxid. Redox Signal. 34, 811-830.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Disbiosis/dietoterapia , Hipertensión/dietoterapia , Inflamación/dietoterapia , Animales , Bacterias/efectos de los fármacos , Bacterias/patogenicidad , Bacteriófagos/genética , Disbiosis/genética , Disbiosis/microbiología , Disbiosis/patología , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Hipertensión/genética , Hipertensión/microbiología , Hipertensión/patología , Inflamación/genética , Inflamación/microbiología , Inflamación/patología
17.
J Reprod Immunol ; 143: 103244, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33186834

RESUMEN

Recent molecular investigations have significantly developed our knowledge of the characteristics of the reproductive microbiome and their associations with host responses to provide an ideal milieu for the development of the embryo during the peri-implantation period and throughout pregnancy as well as to provide a successful in vitro fertilization and appropriate reproductive outcomes. In this context, the establishment of microbial homeostasis in the female reproductive tract, in various physiological periods, is a substantial challenge, which appears the application of probiotics can facilitate the achievement of this goal. So that, currently, probiotics due to its safe and natural features can be considered as a novel biotherapeutic approach. In this review, we comprehensively discuss the bacterial, fungal, and viral diversity detected in the reproductive tract, and their associations with the establishment of dysbiosis/eubiosis conditions as well as we present the significant outcomes on probiotic intervention as an efficient biotherapeutic strategy for management of gestational disorders and improve pregnancy outcomes.


Asunto(s)
Disbiosis/dietoterapia , Genitales Femeninos/microbiología , Microbiota/inmunología , Complicaciones del Embarazo/dietoterapia , Probióticos/uso terapéutico , Suplementos Dietéticos , Disbiosis/inmunología , Disbiosis/microbiología , Femenino , Genitales Femeninos/inmunología , Humanos , Embarazo , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/microbiología , Resultado del Embarazo
18.
Artículo en Inglés | MEDLINE | ID: mdl-33232785

RESUMEN

Emerging evidence highlighted the essential role played by the microbiota-gut-brain axis in maintaining human homeostasis, including nutrition, immunity, and metabolism. Much recent work has linked the gut microbiota to many psychiatric and neurodegenerative disorders such as depression, schizophrenia, and Alzheimer's disease. Shared gut microbiota alterations or dysbiotic microbiota have been identified in these separate disorders relative to controls. Much attention has focused on the bidirectional interplay between the gut microbiota and the brain, establishing gut dysbiotic status as a critical factor in psychiatric disorders. Still, the antibiotic-like effect of psychotropic drugs, medications used for the treatment of these disorders, on gut microbiota is largely neglected. In this review, we summarize the current findings on the impact of psychotropics on gut microbiota and how their antimicrobial potency can trigger dysbiosis. We also discuss the potential therapeutic strategies, including probiotics, prebiotics, and fecal transplantation, to attenuate the dysbiosis related to psychotropics intake.


Asunto(s)
Disbiosis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Terapia Nutricional , Psicotrópicos/efectos adversos , Disbiosis/inducido químicamente , Disbiosis/dietoterapia , Humanos , Terapia Nutricional/métodos
19.
J Nutr Biochem ; 88: 108543, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33144228

RESUMEN

The onset of type 2 diabetes in obesity is associated with gut dysbiosis and a failure to confine commensal bacteria and toxins to the gut lumen while prebiotics may prevent these effects. This study evaluated the effects of pinto beans (PB) supplementation on cecal bacteria, short-chain fatty acids (SCFAs), distal ileal antigen presentation marker (major histocompatibility complex [MHC] II) and antimicrobial peptide genes during short-term high-fat, high sucrose (HFS) feeding. Six-week-old, male C57BL/6J mice were randomly assigned to four groups (n=12/group), and fed a control (C) or HFS diet with or without cooked PB (10%, wt/wt) for 30 days. Supplemental PB in both the C and HFS diets decreased the abundance of Tenericutes and the sulfate-reducing bacteria Bilophila. In contrast, PB raised the abundance of taxa within the SCFAs-producing family, Lachnospiraceae, compared to groups without PB. Consequently, fecal butyric acid was significantly higher in PB-supplemented groups compared to C and HFS groups. PB reversed the HFS-induced ablation of the distal ileal STAT3 phosphorylation, and up-regulated antimicrobial peptide genes (Reg3γ and Reg3ß). Furthermore, the expression of MHC II protein was elevated in the PB supplemented groups compared to C and HFS. Tenericutes and Bilophilia negatively correlated with activated STAT3 and MHC II proteins. Finally, supplemental PB improved fasting blood glucose, glucose tolerance and suppressed TNFα and inducible nitric oxide synthase mRNA in the visceral adipose tissue. Put together, the beneficial impact of PB supplementation on the gut may be central to its potential to protect against diet-induced inflammation and impaired glucose tolerance.


Asunto(s)
Disbiosis/dietoterapia , Microbioma Gastrointestinal , Genes MHC Clase II , Phaseolus , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Animales , Ciego/metabolismo , Dieta Occidental , Suplementos Dietéticos , Disbiosis/metabolismo , Ácidos Grasos Volátiles/metabolismo , Heces/microbiología , Expresión Génica , Humanos , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética
20.
Int J Biol Macromol ; 167: 1587-1597, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33217459

RESUMEN

Undaria pinnatifida was shown to reduce serum lipids and fat accumulation and produce beneficial effect on type 2 diabetes, but its effect on intestinal micro-ecology remains unclear. This study showed that sulfated polysaccharides from U. pinnatifida (UPSP) reduced weight gain, fat accumulation and metabolic disorders in mice fed with high fat diet (HFD). UPSP not only alleviated HFD-induced microbiota dysbiosis indicated as increased abundances of some Bacteroidales members that had positive correlations with the improvement of physiological indexes, but also maintained gut barrier integrity and reduced metabolic endotoxemia. A dose-effect relationship was observed between the dose of UPSP and its effect on some physiological indexes, gut microbiota community and nutrient utilization. The in vitro result showed that the use of Bacteroides species within Bacteroidales on UPSP was species-dependent, and the dose of UPSP affected the growth properties of some Bacteroides species. It implied that UPSP can be considered as prebiotic agent to prevent gut dysbiosis and obesity-related diseases in obese individuals.


Asunto(s)
Disbiosis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/dietoterapia , Síndrome Metabólico/dietoterapia , Polisacáridos/farmacología , Sulfatos/farmacología , Undaria/química , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Fármacos Antiobesidad/farmacología , Bacteroides/efectos de los fármacos , Colon/citología , Colon/efectos de los fármacos , Colon/patología , Dieta Alta en Grasa/efectos adversos , Disbiosis/dietoterapia , Disbiosis/metabolismo , Endotoxemia/dietoterapia , Ácidos Grasos Volátiles/análisis , Heces/microbiología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/farmacología , Polisacáridos/análisis , Polisacáridos/aislamiento & purificación , Prebióticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA