Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 369
Filtrar
1.
Nutrients ; 16(15)2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39125280

RESUMEN

Postbiotics are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit to the host. They range from cell wall fragments to metabolites, bacterial lysates, extracellular vesicles, and short-chain fatty acids (SCFAs). Postbiotics may influence carcinogenesis via a variety of mechanisms. They can promote homeostatic immune responses, reduce inflammation, induce selective cytotoxicity against tumor cells, as well as the enabling the control of tumor cell proliferation and enhancing intestinal epithelial barrier function. Therefore, probiotics can serve as an adjunct strategy in anticancer treatment together with chemotherapy and immunotherapy. Up to now, the only relevant postbiotics used as interventions in oncological patients remain vitamin K molecules, with few phase-II and III trials available. In fact, postbiotics' levels are strictly dependent on the gut microbiota's composition, which may vary between individuals and can be altered under different physiological and pathological conditions. Therefore, the lack of consistent clinical evidence supporting postbiotics' efficacy is due to their poor bioavailability, short half-life, and fluctuating levels. Synbiotics, a mixture of prebiotics and probiotics, are expected to have a more homogeneous bioavailability with respect to postbiotics and may have greater potential for future development. In this review, we focus on the role of postbiotics as an adjuvant therapy in cancer treatment.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias , Probióticos , Humanos , Neoplasias/terapia , Probióticos/uso terapéutico , Prebióticos/administración & dosificación , Simbióticos/administración & dosificación
2.
Commun Biol ; 7(1): 943, 2024 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-39098939

RESUMEN

To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.


Asunto(s)
Bifidobacterium longum subspecies infantis , Ácidos Grasos Volátiles , Leche Humana , Oligosacáridos , Humanos , Leche Humana/metabolismo , Leche Humana/química , Oligosacáridos/metabolismo , Ácidos Grasos Volátiles/metabolismo , Lactante , Bifidobacterium longum subspecies infantis/metabolismo , Bifidobacterium/metabolismo , Probióticos/administración & dosificación , Microbioma Gastrointestinal , Heces/microbiología , Femenino , Simbióticos/administración & dosificación , Preescolar
3.
Adv Exp Med Biol ; 1449: 113-133, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39060734

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major health problem worldwide, and the strongest determinant of liver disease in children. The possible influence of high-fat/low-fiber dietary patterns with microbiota (e.g., increased Firmicutes/Bacteroidetes ratio), and ultimately with MASLD occurrence and progression has been elucidated by several association studies. The possible mechanisms through which microbes exert their detrimental effects on MASLD include gut vascular barrier damage, a shift towards non-tolerogenic immunologic environment, and the detrimental metabolic changes, including a relative reduction of propionate and butyrate in favor of acetate, endogenous ethanol production, and impairment of the unconjugated bile acid-driven FXR-mediated gut-liver axis. The impact of nutritional and probiotic interventions in children with MASLD is described.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Probióticos , Simbióticos , Humanos , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Simbióticos/administración & dosificación , Niño , Trasplante de Microbiota Fecal/métodos , Hígado Graso/terapia , Hígado Graso/microbiología , Hígado Graso/patología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/metabolismo
4.
Food Funct ; 15(15): 7865-7882, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-38967039

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized primarily by cognitive impairment. Recent investigations have highlighted the potential of nutritional interventions that target the gut-brain axis, such as probiotics and prebiotics, in forestalling the onset of AD. In this study, whole-genome sequencing was employed to identify xylan as the optimal carbon source for the tryptophan metabolism regulating probiotic Clostridium sporogenes (C. sporogenes). Subsequent in vivo studies demonstrated that administration of a synbiotic formulation comprising C. sporogenes (1 × 1010 CFU per day) and xylan (1%, w/w) over a duration of 30 days markedly enhanced cognitive performance and spatial memory faculties in the 5xFAD transgenic AD mouse model. The synbiotic treatment significantly reduced amyloid-ß (Aß) accumulation in the cortex and hippocampus of the brain. Importantly, synbiotic therapy substantially restored the synaptic ultrastructure in AD mice and suppressed neuroinflammatory responses. Moreover, the intervention escalated levels of the microbial metabolite indole-3-propionic acid (IPA) and augmented the relative prevalence of IPA-synthesizing bacteria, Lachnospira and Clostridium, while reducing the dominant bacteria in AD, such as Aquabacterium, Corynebacterium, and Romboutsia. Notably, synbiotic treatment also prevented the disruption of gut barrier integrity. Correlation analysis indicated a strong positive association between gut microbiota-generated IPA levels and behavioral changes. In conclusion, this study demonstrates that synbiotic supplementation significantly improves cognitive and intellectual deficits in 5xFAD mice, which could be partly attributed to enhanced IPA production by gut microbiota. These findings provide a theoretical basis for considering synbiotic therapy as a novel microbiota-targeted approach for the treatment of metabolic and neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Clostridium , Disfunción Cognitiva , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Indoles , Ratones Transgénicos , Simbióticos , Xilanos , Animales , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Ratones , Simbióticos/administración & dosificación , Indoles/metabolismo , Disfunción Cognitiva/terapia , Disfunción Cognitiva/metabolismo , Xilanos/metabolismo , Xilanos/farmacología , Clostridium/metabolismo , Masculino , Péptidos beta-Amiloides/metabolismo , Humanos , Propionatos/metabolismo , Eje Cerebro-Intestino/fisiología
5.
Sci Rep ; 14(1): 15471, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969697

RESUMEN

This study examines the effect of phycoerythrin (PE) from a cyanobacterial Nostoc strain encapsulated with alginate as a potential prebiotic to produce synbiotic ice cream products with Lactobacillus casei. It was found that the addition of the encapsulated PE affected, mostly favourably, the physicochemical properties, antioxidant activity, probiotic survival, volatile compound contents, and sensory acceptability of the synbiotic ice cream samples before and after aging at the freezing periods of one day to eight weeks. Thus, it confirms the prebiotic potential of PE for synbiotic ice creams with L. casei.


Asunto(s)
Alginatos , Helados , Lacticaseibacillus casei , Ficoeritrina , Simbióticos , Lacticaseibacillus casei/metabolismo , Helados/microbiología , Alginatos/química , Ficoeritrina/química , Simbióticos/administración & dosificación , Antioxidantes/química , Nostoc/metabolismo , Probióticos
6.
Nutrients ; 16(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38999830

RESUMEN

Insufficient dietary fiber intake can negatively affect the intestinal microbiome and, over time, may result in gut dysbiosis, thus potentially harming overall health. This randomized controlled trial aimed to improve the gut microbiome of individuals with low dietary fiber intake (<25 g/day) during a 7-week synbiotic intervention. The metabolically healthy male participants (n = 117, 32 ± 10 y, BMI 25.66 ± 3.1 kg/m2) were divided into two groups: one receiving a synbiotic supplement (Biotic Junior, MensSana AG, Forchtenberg, Germany) and the other a placebo, without altering their dietary habits or physical activity. These groups were further stratified by their dietary fiber intake into a low fiber group (LFG) and a high fiber group (HFG). Stool samples for microbiome analysis were collected before and after intervention. Statistical analysis was performed using linear mixed effects and partial least squares models. At baseline, the microbiomes of the LFG and HFG were partially separated. After seven weeks of intervention, the abundance of SCFA-producing microbes significantly increased in the LFG, which is known to improve gut health; however, this effect was less pronounced in the HFG. Beneficial effects on the gut microbiome in participants with low fiber intake may be achieved using synbiotics, demonstrating the importance of personalized synbiotics.


Asunto(s)
Fibras de la Dieta , Heces , Microbioma Gastrointestinal , Simbióticos , Humanos , Simbióticos/administración & dosificación , Masculino , Fibras de la Dieta/administración & dosificación , Adulto , Método Doble Ciego , Heces/microbiología , Adulto Joven
7.
Front Immunol ; 15: 1418594, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975337

RESUMEN

Introduction: Maternal synbiotic supplementation during pregnancy and lactation can significantly influence the immune system. Prebiotics and probiotics have a positive impact on the immune system by preventing or ameliorating among others intestinal disorders. This study focused on the immunomodulatory effects of B. breve M-16V and short chain galacto-oligosaccharides (scGOS)/long chain fructo-oligosachairdes (lcFOS), including systemic and mucosal compartments and milk composition. Methods: Lewis rats were orally administered with the synbiotic or vehicle during pregnancy (21 days) and lactation (21 days). At the weaning day, small intestine (SI), mammary gland (MG), adipose tissue, milk, mesenteric lymph nodes (MLN), salivary gland (SG), feces and cecal content were collected from the mothers. Results: The immunoglobulinome profile showed increased IgG2c in plasma and milk, as well as elevated sIgA in feces at weaning. The supplementation improved lipid metabolism through enhanced brown adipose tissue activity and reinforced the intestinal barrier by increasing the expression of Muc3, Cldn4, and Ocln. The higher production of short chain fatty acids in the cecum and increased Bifidobacterium counts suggest a potential positive impact on the gastrointestinal tract. Discussion: These findings indicate that maternal synbiotic supplementation during gestation and lactation improves their immunological status and improved milk composition.


Asunto(s)
Bifidobacterium breve , Lactancia , Leche , Oligosacáridos , Animales , Femenino , Embarazo , Bifidobacterium breve/inmunología , Leche/inmunología , Leche/química , Ratas , Ratas Endogámicas Lew , Suplementos Dietéticos , Simbióticos/administración & dosificación , Probióticos/administración & dosificación , Probióticos/farmacología
9.
Nutrients ; 16(13)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38999862

RESUMEN

Irritable bowel syndrome (IBS) is a common gastrointestinal disorder with gut microbiota imbalance playing a significant role. There are increasing numbers of research studies exploring treatment options involving probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT), but it is still uncertain which treatment option is superior. The research was conducted on various databases and unpublished trial data (up to February 2023). Randomized controlled trials (RCTs) were screened for adult patients with IBS comparing interventions with placebo. Probiotics, prebiotics, synbiotics, and FMT were assessed for their impact using mean difference and Bayesian network meta-analysis. Out of 6528 articles, 54 were included for probiotics, 7 for prebiotics/synbiotics, and 6 for FMT. Probiotics showed improvement in IBS symptoms, particularly with Bifidobacterium and Lactobacillus strains. Prebiotics and synbiotics did not show significant improvement. Network meta-analysis indicated the favorable effects of probiotics (OR = 0.53, 95% CI, 0.48 to 0.59) and FMT (OR = 0.46, 95% CI, 0.33 to 0.64) on IBS, with no serious adverse events reported. In short, probiotics and FMT are effective for managing IBS, with Bifidobacterium and Lactobacillus being dominant strains. However, the most effective probiotic combination or strain remains unclear, while prebiotics and synbiotics did not show significant improvement.


Asunto(s)
Trasplante de Microbiota Fecal , Síndrome del Colon Irritable , Metaanálisis en Red , Prebióticos , Probióticos , Simbióticos , Síndrome del Colon Irritable/terapia , Síndrome del Colon Irritable/microbiología , Humanos , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Simbióticos/administración & dosificación , Resultado del Tratamiento , Microbioma Gastrointestinal , Ensayos Clínicos Controlados Aleatorios como Asunto , Bifidobacterium , Adulto , Femenino , Lactobacillus , Masculino
10.
Brain Behav Immun ; 120: 360-371, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38885746

RESUMEN

Irritability worsens prognosis and increases mortality in individuals with Attention-Deficit and Hyperactivity Disorder (ADHD) and/or Borderline Personality Disorder (BPD). However, treatment options are still insufficient. The aim of this randomized, double blind, placebo-controlled study was to investigate the superiority of a synbiotic over placebo in the management of adults with ADHD and/or BPD and high levels of irritability. The study was conducted between February 2019 and October 2020 at three European clinical centers located in Hungary, Spain and Germany. Included were patients aged 18-65 years old diagnosed with ADHD and/or BPD and high levels of irritability (i.e., an Affectivity Reactivity Index (ARI-S) ≥ 5, plus a Clinical Global Impression-Severity Scale (CGI-S) score ≥ 4). Subjects were randomized 1(synbiotic):1(placebo); the agent was administered each day, for 10 consecutive weeks. The primary outcome measure was end-of-treatment response (i.e., a reduction ≥ 30 % in the ARI-S total score compared to baseline, plus a Clinical Global Impression-Improvement (CGI-I) total score of < 3 (very much, or much improved) at week 10). Between-treatment differences in secondary outcomes, as well as safety were also investigated. Of the 231 included participants, 180 (90:90) were randomized and included in the intention-to-treat-analyses. Of these, 117 (65 %) were females, the mean age was 38 years, ADHD was diagnosed in 113 (63 %), BPD in 44 (24 %), both in 23 (13 %). The synbiotic was well tolerated. At week 10, patients allocated to the synbiotic experienced a significantly higher response rate compared to those allocated to placebo (OR: 0.2, 95 % CI:0.1 to 0.7; P = 0.01). These findings suggest that that (add-on) treatment with a synbiotic may be associated with a clinically meaningful improvement in irritability in, at least, a subgroup of adults with ADHD and/or BPD. A superiority of the synbiotic over placebo in the management of emotional dysregulation (-3.6, 95 % CI:-6.8 to -0.3; P = 0.03), emotional symptoms (-0.6, 95 % CI:-1.2 to -0.05; P = 0.03), inattention (-1.8, 95 % CI: -3.2 to -0.4; P = 0.01), functioning (-2.7, 95 % CI: -5.2 to -0.2; P = 0.03) and perceived stress levels (-0.6, 95 % CI: -1.2 to -0.05; P = 0.03) was also suggested. Higher baseline RANK-L protein levels were associated with a significantly lower response rate, but only in the synbiotic group (OR: 0.1, 95 % CI: -4.3 to - 0.3, P = 0.02). In the placebo group, higher IL-17A levels at baseline were significantly associated with a higher improvement in in particular, emotional dysregulation (P = 0.04), opening a door for new (targeted) drug intervention. However, larger prospective studies are warranted to confirm the findings. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03495375.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno de Personalidad Limítrofe , Genio Irritable , Simbióticos , Humanos , Adulto , Masculino , Femenino , Trastorno por Déficit de Atención con Hiperactividad/terapia , Trastorno de Personalidad Limítrofe/terapia , Trastorno de Personalidad Limítrofe/psicología , Persona de Mediana Edad , Simbióticos/administración & dosificación , Método Doble Ciego , Resultado del Tratamiento , Adulto Joven , Adolescente , Anciano , España , Alemania
11.
Microbiol Res ; 286: 127787, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38851010

RESUMEN

Vaginal health is essential to a woman's overall well-being, as abnormalities in vaginal health can lead to a variety of gynaecological disorders, such as urinary tract infections, yeast infections, and bacterial vaginosis. The vaginal microbiome is essential for the prevention of these infections. Disruptions in this microbial ecosystem can significantly impact vaginal health. The concept of utilizing probiotics and prebiotics to stimulate the growth of protective vaginal microbiota has gathered substantial interest in recent years. Probiotics are live micro-organisms that strengthen and restore vaginal microbial balance by lowering pH levels, production of bacteriocins, biofilm disruption, modulation of immune response, and production of hydrogen peroxide (H2O2), consequently combating the development of pathogens. Prebiotics are oligosaccharides that encourage the development of probiotics such as lactobacilli species. Probiotics and prebiotics also have some broader implications for vaginal health, including their role in minimizing the incidence of premature birth, optimizing fertility, managing menopausal symptoms, and preventing vaginal infections. Synbiotics are a combination of probiotics and prebiotics that deliver additional benefits by encouraging the development and activity of beneficial microbes. Furthermore, postbiotics are bioactive compounds derived from probiotic bacteria during fermentation that have immunomodulatory actions and provide an additional layer of protection against vaginal infections. The present study highlights the most prevalent vaginal infections and limitations of existing therapies that influence the vaginal microbiota. The profound consequences of probiotics and prebiotics in women's health, including their role in minimizing the prevalence of vaginal infections and promoting overall vaginal health, as well as advanced therapeutic strategies such as synbiotics and postbiotics, are also discussed. The literature offers significant insights into the mechanism, efficacy, and safety of probiotics and prebiotics to healthcare providers and researchers.


Asunto(s)
Microbiota , Prebióticos , Probióticos , Vagina , Humanos , Femenino , Probióticos/administración & dosificación , Vagina/microbiología , Vaginosis Bacteriana/prevención & control , Vaginosis Bacteriana/microbiología , Vaginosis Bacteriana/terapia , Lactobacillus , Simbióticos/administración & dosificación
12.
Clin Res Hepatol Gastroenterol ; 48(7): 102397, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38879003

RESUMEN

BACKGROUND: Metabolic dysfunction-associated steatotic liver disease (MASLD) is constantly rising globally. There are barely any effective medications or supplements for the management of MASLD. We aim to systematically evaluate the most current evidence for gut microbiota-regulating supplements in patients with MASLD. METHODS: We searched multiple electronic data for randomized controlled trials (RCTs) published from January 1, 2012, to July 15, 2023. The intervention measures included probiotics, prebiotics, synbiotics, antibiotics, and fecal microbiota transplantation (FMT). The control group was treated with a placebo or usual care. The intervention duration was divided into two periods (>12 weeks and ≤12 weeks). Adequate evaluation data for antibiotics and FMT have not been obtained. Therefore, the other three microbiota regulators are the primary evaluation measures in this study. RESULTS: We found that probiotics alone could not improve clinical indicators in MASLD patients. However, synbiotics exhibited an improvement in reducing liver steatosis, TNF-ɑ levels, and increasing HDL-c levels, and the inflammatory markers of liver cells (ALT and AST) were also improved. For the effective intervention duration, this systematic review suggested that around 12 weeks is an ideal intervention cycle for MASLD patients. CONCLUSIONS: This meta-analysis supported the modulation of gut microbiota with synbiotics in the management of MASLD.


Asunto(s)
Hígado Graso , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Prebióticos , Probióticos , Simbióticos , Humanos , Antibacterianos/administración & dosificación , Hígado Graso/terapia , Microbioma Gastrointestinal/fisiología , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Simbióticos/administración & dosificación
13.
Biochem Biophys Res Commun ; 726: 150274, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-38924882

RESUMEN

Alzheimer's disease (AD) is a complex neurodegenerative condition with growing evidence implicating the gut microbiota in its pathogenesis. This study aimed to investigate the effects of NMN synbiotics, a combination of ß-nicotinamide mononucleotide (NMN), Lactobacillus plantarum, and lactulose, on the gut microbiota composition and metabolic profiles in APP/PS1 transgenic mice. Results demonstrated that NMN synbiotics led to a notable restructuring of the gut microbiota, with a decreased Firmicutes/Bacteroidetes ratio in the AD mice, suggesting a potential amelioration of gut dysbiosis. Alpha diversity indices indicated a reduction in microbial diversity following NMN synbiotics supplementation, while beta diversity analyses revealed a shift towards a more balanced microbial community structure. Functional predictions based on the 16S rRNA data highlighted alterations in metabolic pathways, particularly those related to amino acid and energy metabolism, which are crucial for neuronal health. The metabolomic analysis uncovered a significant impact of NMN synbiotics on the gut metabolome, with normalization of metabolic composition in AD mice. Differential metabolite functions were enriched in pathways associated with neurotransmitter synthesis and energy metabolism, pointing to the potential therapeutic effects of NMN synbiotics in modulating the gut-brain axis and synaptic function in AD. Immunohistochemical staining observed a significant reduction of amyloid plaques formed by Aß deposition in the brain of AD mice after NMN synbiotics intervention. The findings underscore the potential of using synbiotics to ameliorate the neurodegenerative processes associated with Alzheimer's disease, opening new avenues for therapeutic interventions.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Ratones Transgénicos , Simbióticos , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/dietoterapia , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/microbiología , Simbióticos/administración & dosificación , Ratones , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Presenilina-1/metabolismo , Presenilina-1/genética , Mononucleótido de Nicotinamida/metabolismo , Masculino , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/dietoterapia , Disbiosis/terapia
14.
Food Chem Toxicol ; 190: 114843, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38944142

RESUMEN

Mulberry (Morus alba L) fruit is traditionally used in Chinese medicine and has several beneficial effects, such as hypoglycemic, hypolipidemic, and anti-oxidative effects. We previously developed the synbiotic mulberry (SM) containing probiotic Lactobacilli, prebiotic inulin, and mulberry powder. In food supplement development, toxicity is the most important criterion in food and drug regulations before commercialization. Thus, this study aimed to investigate the subchronic toxicity of SM in male and female Wistar rats to evaluate its biosafety. The subchronic toxicity study was conducted by daily oral administration of SM at doses of 250, 500, and 1000 mg/kgBW for 90 days. Male and female rats were evaluated for body weight, organ coefficients, biochemical and hematological parameters, and vital organ histology. The results showed no mortality or toxic changes in the subchronic toxicity study. These results suggested that no observed adverse effect level (NOAEL) of SM in male and female rats has been considered at 1000 mg/kgBW for subchronic toxicity study.


Asunto(s)
Morus , Simbióticos , Animales , Femenino , Masculino , Ratas , Administración Oral , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Morus/química , Nivel sin Efectos Adversos Observados , Tamaño de los Órganos/efectos de los fármacos , Ratas Wistar , Simbióticos/administración & dosificación , Pruebas de Toxicidad Subcrónica
15.
Cell Host Microbe ; 32(7): 1163-1176.e6, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38906158

RESUMEN

Depletion of beneficial microbes by modern lifestyle factors correlates with the rising prevalence of food allergies. Re-introduction of allergy-protective bacteria may be an effective treatment strategy. We characterized the fecal microbiota of healthy and food-allergic infants and found that the anaerobe Anaerostipes caccae (A. caccae) was representative of the protective capacity of the healthy microbiota. We isolated a strain of A. caccae from the feces of a healthy infant and identified lactulose as a prebiotic to optimize butyrate production by A. caccae in vitro. Administration of a synbiotic composed of our isolated A. caccae strain and lactulose increased luminal butyrate in gnotobiotic mice colonized with feces from an allergic infant and in antibiotic-treated specific pathogen-free (SPF) mice, and prevented or treated an anaphylactic response to allergen challenge. The synbiotic's efficacy in two models and microbial contexts suggests that it may be a promising approach for the treatment of food allergy.


Asunto(s)
Heces , Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Lactulosa , Simbióticos , Animales , Simbióticos/administración & dosificación , Hipersensibilidad a los Alimentos/prevención & control , Ratones , Humanos , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Lactante , Butiratos/metabolismo , Prebióticos/administración & dosificación , Femenino , Modelos Animales de Enfermedad , Organismos Libres de Patógenos Específicos , Vida Libre de Gérmenes , Masculino
16.
Nutrients ; 16(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38892479

RESUMEN

Myalgic encephalomyelitis, also known as chronic fatigue syndrome (ME/CFS), and long COVID are complex, multisystemic and long-term disabling conditions characterized by debilitating post-exertional malaise and other core symptoms related to immune dysregulation resultant from post-viral infection, including mitochondrial dysfunction, chronic neuroinflammation and gut dysbiosis. The reported associations between altered microbiota composition and cardinal symptoms of ME/CFS and long COVID suggest that the use of microbial preparations, such as probiotics, by restoring the homeostasis of the brain-immune-gut axis, may help in the management of symptoms in both conditions. Therefore, this review aims to investigate the implications of alerted gut microbiome and assess the evidence supporting use of microbial-based preparations, including probiotics, synbiotics, postbiotics alone and/or in combination with other nutraceuticals in the management of fatigue, inflammation and neuropsychiatric and gastrointestinal symptoms among patients with ME/CFS and long COVID.


Asunto(s)
COVID-19 , Disbiosis , Síndrome de Fatiga Crónica , Microbioma Gastrointestinal , Probióticos , Humanos , Síndrome de Fatiga Crónica/terapia , COVID-19/complicaciones , COVID-19/inmunología , Probióticos/uso terapéutico , SARS-CoV-2 , Suplementos Dietéticos , Simbióticos/administración & dosificación , Eje Cerebro-Intestino
17.
Nutrients ; 16(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38931246

RESUMEN

Immune system development during gestation and suckling is significantly modulated by maternal environmental and dietary factors. Breastfeeding is widely recognized as the optimal source of nutrition for infant growth and immune maturation, and its composition can be modulated by the maternal diet. In the present work, we investigated whether oral supplementation with Bifidobacterium breve M-16V and short-chain galacto-oligosaccharide (scGOS) and long-chain fructo-oligosaccharide (lcFOS) to rat dams during gestation and lactation has an impact on the immune system and microbiota composition of the offspring at day 21 of life. On that day, blood, adipose tissue, small intestine (SI), mesenteric lymph nodes (MLN), salivary gland (SG), cecum, and spleen were collected. Synbiotic supplementation did not affect the overall body or organ growth of the pups. The gene expression of Tlr9, Muc2, IgA, and Blimp1 were upregulated in the SI, and the increase in IgA gene expression was further confirmed at the protein level in the gut wash. Synbiotic supplementation also positively impacted the microbiota composition in both the small and large intestines, resulting in higher proportions of Bifidobacterium genus, among others. In addition, there was an increase in butanoic, isobutanoic, and acetic acid concentrations in the cecum but a reduction in the small intestine. At the systemic level, synbiotic supplementation resulted in higher levels of immunoglobulin IgG2c in plasma, SG, and MLN, but it did not modify the main lymphocyte subsets in the spleen and MLN. Overall, synbiotic maternal supplementation is able to positively influence the immune system development and microbiota of the suckling offspring, particularly at the gastrointestinal level.


Asunto(s)
Animales Lactantes , Bifidobacterium breve , Suplementos Dietéticos , Microbioma Gastrointestinal , Oligosacáridos , Simbióticos , Animales , Simbióticos/administración & dosificación , Femenino , Embarazo , Ratas , Fenómenos Fisiologicos Nutricionales Maternos , Lactancia , Sistema Inmunológico , Masculino , Animales Recién Nacidos
18.
Animal ; 18(6): 101185, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38843664

RESUMEN

Although anticoccidials effectively control coccidiosis, a needed reduction in the reliance on antimicrobials in animal production leads to the exploration of alternative compounds. The present study aimed to test five different dietary treatments to counteract the negative impact of coccidiosis on broiler chickens' health and performance. 1-day-old male Ross 308 broilers (n = 960) were randomly assigned to one of eight treatments, with six cages per treatment (20 birds/cage). To the diet of the broiler chickens of treatments (Trt) 1-5, a synbiotic was added from d0-10. From d10-28, birds of Trt1 and Trt2 were fed synbiotics, whereas birds of Trt3 were fed diets with glutamine, and birds of Trt4 and Trt5 were fed diets with a combination of ß-glucans and betaine. From d28-35 onwards, birds of Trt1 were fed a diet with a synbiotic, whereas birds of Trt2-4 received diets with glutamine, and birds of Trt5 were fed a non-supplemented diet. Birds of the positive control group (PC; Trt6) were fed a standard diet supplemented with an anticoccidial (Decoquinate). The challenged negative control (NCchall; Trt7) and non-challenged negative control (NC) Trt8 were fed a standard diet without anticoccidial or other dietary treatment. At 7 days (d) of age, all birds were inoculated with 1 023, 115, and 512 sporulated oocysts of E. acervulina, E. maxima, and E. tenella, respectively, except for Trt8. Body weight gain (BWG), feed intake, and feed conversion ratio were assessed for each feeding phase (d0-10, d10-28 and d28-35) and overall experimental period (d0-35). Oocyst shedding, Eimeria lesion scores, cecal length, and relative weight were assessed at d13, d22, d28 and d35. Additionally, oocyst shedding was determined at d9 and d17. Litter quality was evaluated at d27 and d34, and footpad lesions at d34. During the starter (d0-10) and finisher (d28-35) periods, performance did not differ between the treatments. During the grower period (d10-28), Trt6 (PC) and Trt8 (NC) chickens had the highest BWG of all treatments (P < 0.001). Dietary treatment had no effect on litter quality and severity of footpad lesions. In the PC group (Trt6), low oocyst excretion and lesion scores were found. When comparing Trt1-5 with NCchall (Trt7), none of the treatments significantly reduced oocyst output or lesion scores. In conclusion, in this experiment, none of the dietary treatments performed similar or better compared to the PC group (Trt6) regarding performance or reducing Eimeria oocyst shedding or lesion scores.


Asunto(s)
Alimentación Animal , Pollos , Coccidiosis , Dieta , Eimeria , Oocistos , Enfermedades de las Aves de Corral , Animales , Coccidiosis/veterinaria , Enfermedades de las Aves de Corral/parasitología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/tratamiento farmacológico , Masculino , Alimentación Animal/análisis , Eimeria/fisiología , Dieta/veterinaria , Suplementos Dietéticos/análisis , Simbióticos/administración & dosificación , Distribución Aleatoria , Betaína/administración & dosificación , Betaína/farmacología , Glutamina/farmacología , Glutamina/administración & dosificación , beta-Glucanos/farmacología , beta-Glucanos/administración & dosificación , beta-Glucanos/uso terapéutico
19.
Nutrients ; 16(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38892536

RESUMEN

The diversity and functionality of gut microbiota may play a crucial role in the function of human motor-related systems. In addition to traditional nutritional supplements, there is growing interest in microecologics due to their potential to enhance sports performance and facilitate post-exercise recovery by modulating the gut microecological environment. However, there is a lack of relevant reviews on this topic. This review provides a comprehensive overview of studies investigating the effects of various types of microecologics, such as probiotics, prebiotics, synbiotics, and postbiotics, on enhancing sports performance and facilitating post-exercise recovery by regulating energy metabolism, mitigating oxidative-stress-induced damage, modulating immune responses, and attenuating bone loss. Although further investigations are warranted to elucidate the underlying mechanisms through which microecologics exert their effects. In summary, this study aims to provide scientific evidence for the future development of microecologics in athletics.


Asunto(s)
Atletas , Rendimiento Atlético , Ejercicio Físico , Microbioma Gastrointestinal , Probióticos , Humanos , Rendimiento Atlético/fisiología , Probióticos/administración & dosificación , Microbioma Gastrointestinal/fisiología , Ejercicio Físico/fisiología , Prebióticos/administración & dosificación , Simbióticos/administración & dosificación , Metabolismo Energético , Estrés Oxidativo , Suplementos Dietéticos , Recuperación Después del Ejercicio
20.
Nutrients ; 16(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892608

RESUMEN

Gut microbiome-modulating agents (MMAs), including probiotics, prebiotics, postbiotics, and synbiotics, are shown to ameliorate type 1 diabetes (T1D) by restoring the microbiome from dysbiosis. The objective of this systematic review and meta-analysis was to assess the impact of MMAs on hemoglobin A1c (HbA1c) and biomarkers associated with (T1D). A comprehensive search was conducted in PubMed, Web of Science, Embase, Cochrane Library, National Knowledge Infrastructure, WeiPu, and WanFang Data up to 30 November 2023. Ten randomized controlled trials (n = 630) were included, with study quality evaluated using the Cochrane risk-of-bias tool. Random-effect models with standardized mean differences (SMDs) were utilized. MMA supplementation was associated with improvements in HbA1c (SMD = -0.52, 95% CI [-0.83, -0.20]), daily insulin usage (SMD = -0.41, 95% confidence interval (CI) [-0.76, -0.07]), and fasting C-peptide (SMD = 0.99, 95% CI [0.17, 1.81]) but had no effects on FBG, CRP, TNF-α, IL-10, LDL, HDL, and the Shannon index. Subgroup analysis of HbA1c indicated that a long-term intervention (>3 months) might exert a more substantial effect. These findings suggest an association between MMAs and glycemic control in T1D. Further large-scale clinical trials are necessary to confirm these findings with investigations on inflammation and gut microbiota composition while adjusting confounding factors such as diet, physical activity, and the dose and form of MMA intervention.


Asunto(s)
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Hemoglobina Glucada , Probióticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/sangre , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Hemoglobina Glucada/metabolismo , Probióticos/uso terapéutico , Prebióticos/administración & dosificación , Biomarcadores/sangre , Simbióticos/administración & dosificación , Suplementos Dietéticos , Femenino , Disbiosis , Adulto , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA