Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 652
Filtrar
1.
Cureus ; 16(9): e68404, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39360063

RESUMEN

The human gut is the abode of several complex and diverse microbes. It is a fact that the human brain is interconnected with the spinal cord and sense organs; however, there is also a possibility of a connection between the brain and the gut microbiome. The human gut can be altered in various ways, the principal method being the intake of prebiotics, probiotics and synbiotics. Can this alteration in the gut microbiome be clinically utilised in the perioperative period? We conducted a literature search related to this topic using databases and search engines (Medical Literature Analysis and Retrieval System Online {MEDLINE}, Embase, Scopus, PubMed and Google Scholar). The search revealed some preclinical and clinical studies in animals and humans that demonstrate the alteration of the gut microbiome with the use of anxiolysis, probiotics/prebiotics and other perioperative factors including opioids, anaesthetics and perioperative stress. The significant effects of this alteration have been seen on preoperative anxiety and postoperative delirium/cognitive dysfunction/pain. These effects are described in this narrative review, which opens up newer vistas for high-quality research related to the gut microbiome, gut-brain axis, the related signaling pathways and their clinical application in the perioperative period.

2.
Ann N Y Acad Sci ; 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39392836

RESUMEN

Over the last two decades, advancements in sequencing technologies have significantly deepened our understanding of the human microbiome's complexity, leading to increased concerns about the detrimental effects of antibiotics on these intricate microbial ecosystems. Concurrently, the rise in antimicrobial resistance has intensified the focus on how beneficial microbes can be harnessed to treat diseases and improve health and offer potentially promising alternatives to traditional antibiotic treatments. Here, we provide a comprehensive overview of both established and emerging microbe-centric therapies, from probiotics to advanced microbial ecosystem therapeutics, examine the sophisticated ways in which microbes are used medicinally, and consider their impacts on microbiome homeostasis and health outcomes through a microbial ecology lens. In addition, we explore the concept of rewilding the human microbiome by reintroducing "missing microbes" from nonindustrialized societies and personalizing microbiome modulation to fit individual microbial profiles-highlighting several promising directions for future research. Ultimately, the advancements in sequencing technologies combined with innovative microbial therapies and personalized approaches herald a new era in medicine poised to address antibiotic resistance and improve health outcomes through targeted microbiome management.

3.
Nutr Health ; : 2601060241282357, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39370911

RESUMEN

Background: Stunting is among the main obstacles to human development affecting millions of children worldwide, particularly in the sub-Saharan Africa region. Randomized clinical trials have shown the positive effects of prebiotics, probiotics, and synbiotics in improving growth in children and toddlers. However, although the global mobilization to tackle its challenges in their different aspects is visible, it remains to define effective large-scale up interventions and strategies to obtain long-lasting impacts. Objective: The objective of this review is to re-evaluate the efficacy of prebiotics, probiotics, and/or synbiotics on growth in children 0 to 5 years in Africa including recently published studies. Methods: Systematic search will be carried out in Pubmed, Science Direct, clinicaltrial.org, and Google Scholar. Both randomized and observational studies that assess the association between prebiotics, probiotics, and synbiotics, and health benefits and growth in children under 5 years of age will be included in the review. PRISMA-P (preferred reporting items for systematic review and meta-analysis protocols) will be used used for this protocol, and PRISMA will be used for the systematic review. The Cochrane Risk Assessment Tool will be used to assess the quality of eligible studies. If the compiled data are appropriate and sufficient enough, we will perform a meta-analysis using RevMan software. Conclusion: This review will provide up-to-date and reliable information on the efficacy of prebiotics, probiotics, and synbiotics on the growth of children under 5 years of age especially in developing countries. PROSPERO registration number CRD42022343138.

4.
Heliyon ; 10(16): e36157, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39247358

RESUMEN

Background: Hypertension is a global public health concern. A synbiotic preparation containing Bifidobacterium lactis and Lactobacillus acidophilus has been used as adjunct therapy for hypertension. We sought to elucidate the antihypertensive activity of this preparation and explore the underlying mechanisms. Methods and results: Blood pressure in rats was measured using the tail-cuff method. Colonization of the gastrointestinal tract by the two probiotics was determined by real-time quantitative polymerase chain reaction (qPCR). Mechanistic studies were performed by proteomic analyses based on liquid chromatography-mass spectrometry and STRING database and metabolomic analyses using the UHPLC-Q-TOF/MS platform and peroxisome proliferator-activated receptor (PPAR)ß/γ antagonists. Although biochemical analysis of blood samples showed that the synbiotic preparation did not alter the levels of angiotensin II, aldosterone, or cortisol, it significantly lowered the systolic blood pressure in the treatment group. Moreover, the synbiotic preparation contributed to the localization of the two probiotics in the ileum and colon of the treatment group. Proteomics, immunochemistry, and real-time qPCR analyses showed that administration of the synbiotic preparation activated the PPAR signaling pathway in the ileum and significantly upregulated PPARß and PPARγ. The antagonist studies further confirmed this finding. In addition, metabolomic analyses demonstrated that among the 27 metabolites that showed significant differences between the control and model groups, administration of the synbiotic preparation significantly upregulated lysophosphatidylethanolamine and phosphatidylcholine in the ileum of the treatment group. Conclusion: The results of the study suggest that the novel synbiotic preparation reduces blood pressure by altering the composition of the intestinal microbiota, regulating PPAR signaling pathway, and activating the PPARß and PPARγ cascade reactions in the ileum.

5.
Environ Int ; 192: 109017, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39317009

RESUMEN

Human exposure to metal(loid)s has dramatically increased over the past five decades, which has triggered public concern worldwide. Recently, gut microbiota has been considered a target for metal(loid)s, and some literature has reviewed the interactions between gut microbiota and heavy metal(loid)s (HMs) with high toxicity. However, whether there is an interaction between gut microbiota and metal(loid)s with essential roles or some normal functions are far from clear to date. Importantly, in addition to traditional probiotics that have been clarified to alleviate the adverse effect of HMs on the body, some novel probiotics, prebiotics, synbiotics, and postbiotics may also exhibit comparable or even better abilities of metal(loid) remediation. In this review, we mainly outline and discuss recent research findings on the metal(loid)-gut microbiota interactions and microbiota-related protective strategies.

6.
World J Diabetes ; 15(9): 1833-1836, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39280175

RESUMEN

The gut microbiota is important in the development and progression of metabolic illnesses such type 2 diabetes, cardiovascular disease (CVD), and obesity. This diverse community of microorganisms controls a variety of physiological functions, including metabolism, inflammation, and immune response. Understanding these interactions has resulted in novel therapeutic options, including microbiome supplementation. The gut microbiome is extremely susceptible to dietary changes, which can alter its makeup and function, influencing metabolite synthesis that affects host health. Certain metabolites, such as butyrate and propionate, have been proven to protect against metabolic illnesses, whereas trimethylamine has been linked to CVD. Prebiotics, probiotics, synbiotics, and postbiotics are being investigated by researchers as ways to change the gut microbiome and boost metabolic health. Despite advances in therapy and lifestyle adjustments, the prevalence of metabolic syndrome is increasing, emphasizing the need for new medicines.

7.
Heliyon ; 10(16): e35980, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39229543

RESUMEN

Over recent years, the scientific community has acknowledged the crucial role of certain microbial strains inhabiting the intestinal ecosystem in promoting human health, and participating in various beneficial functions for the host. These microorganisms are now referred to as next-generation probiotics and are currently considered as biotherapeutic products and food or nutraceutical supplements. However, the majority of next-generation probiotic candidates pose nutritional demands and exhibit high sensitivity towards aerobic conditions, leading to numerous technological hurdles in large-scale production. This underscores the need for the development of suitable delivery systems capable of enhancing the viability and functionality of these probiotic strains. Currently, potential candidates for next generation probiotics (NGP) are being sought among gut bacteria linked to health, which include strains from the genera Bacteroids, Faecalibacterium, Akkermansia and Clostridium. In contrast to Lactobacillus spp. and Bifidobacterium spp., NGP, particularly Bacteroids spp. and Clostridium spp., appear to exhibit greater ambiguity regarding their potential to induce infectious diseases. The present review provides a comprehensive overview of NGPs in terms of their health beneficial effects, regulation framework and risk assessment targeting relevant criteria for commercialization in food and pharmaceutical markets.

8.
Trials ; 25(1): 615, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289685

RESUMEN

BACKGROUND: Prematurity remains one of the main causes of neonatal morbidity and mortality. Approximately two thirds of preterm births are spontaneous, i.e. secondary to preterm labour, preterm prelabour rupture of membranes (PPROM) or cervical insufficiency. Etiologically, the vaginal microbiome plays an important role in spontaneous preterm birth (sPTB). Vaginal dysbiosis and bacterial vaginosis are well-known risk factors for ascending lower genital tract infections and sPTB, while a Lactobacillus crispatus-dominated vaginal microbiome is associated with term deliveries. Synbiotics may help to achieve and/or maintain a normal, Lactobacillus-dominated vaginal microbiome. METHODS: We will perform a multi-centre, double-blind, randomised, placebo-controlled trial. Women aged 18 years or older with a singleton pregnancy are eligible for inclusion at 80/7-106/7 weeks gestational age if they have one or more of the following risk factors for sPTB: previous sPTB at 240/7-356/7 weeks, prior PPROM before 360/7 weeks, or spontaneous pregnancy loss at 140/7-236/7 weeks of gestation. Exclusion criteria are multiple gestation, cervix conisation, inflammatory bowel disease, uterine anomaly, and the use of pro-/pre-/synbiotics. Patients will be randomised to oral synbiotics or placebo, starting before 11 weeks of gestation until delivery. The oral synbiotic consists of eight Lactobacillus species (including L. crispatus) and prebiotics. The primary outcome is the gestational age at delivery. Vaginal microbiome analysis once per trimester (at approximately 9, 20, and 30 weeks) and delivery will be performed using metataxonomic sequencing (16S rRNA gene) and microbial culture. Secondary outcomes include PPROM, the use of antibiotics, antenatal admission information, and neonatal outcomes. DISCUSSION: This study will evaluate the effect of oral synbiotics on the vaginal microbiome during pregnancy in a high-risk population and correlate the microbial changes with the gestational age at delivery and relevant pregnancy outcomes. TRIAL REGISTRATION: ClinicalTrials.gov, NCT05966649. Registered on April 5, 2024.


Asunto(s)
Estudios Multicéntricos como Asunto , Nacimiento Prematuro , Ensayos Clínicos Controlados Aleatorios como Asunto , Simbióticos , Vagina , Humanos , Femenino , Método Doble Ciego , Embarazo , Nacimiento Prematuro/prevención & control , Simbióticos/administración & dosificación , Vagina/microbiología , Factores de Riesgo , Microbiota , Edad Gestacional , Recién Nacido , Resultado del Tratamiento , Vaginosis Bacteriana/microbiología , Vaginosis Bacteriana/diagnóstico
9.
Cureus ; 16(8): e66875, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39280360

RESUMEN

Chronic obstructive pulmonary disease (COPD) represents a significant global health burden, characterized by progressive airflow limitation and exacerbations that significantly impact patient morbidity and mortality. Recent research has investigated the interplay between the gut and the lungs, known as the gut-lung axis, highlighting the role of the gut microbiome in COPD pathogenesis. Dysbiosis, characterized by microbial imbalance, has implications for COPD, influencing disease progression and susceptibility to exacerbations. This comprehensive review integrates current scientific literature on gut microbiota modulation as a therapeutic avenue for COPD management. Through a thorough discussion of studies investigating probiotics, prebiotics, synbiotics, antibiotics, dietary fiber, and fecal microbiota transplantation, this review summarizes the influence of these interventions on COPD via the gut-lung axis through the modulation of systemic inflammation, mucosal immunity, and metabolic processes. The interventions highlighted here show potential in preventing COPD exacerbations, preserving lung function, and improving patient quality of life. By compiling the latest scientific evidence, this review provides a comprehensive framework for physicians and researchers to deduce the effectiveness of gut microbiome modulation as an adjunctive therapeutic strategy in COPD management.

10.
Pharmacol Res ; 208: 107398, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39241935

RESUMEN

BACKGROUND: Patients with non-alcoholic fatty liver disease (NAFLD) benefit from using synbiotics. However, findings from existing trials remain contentious. Therefore, this meta-analysis evaluated the effects of synbiotics on liver enzymes, blood pressure, inflammation, and lipid profiles in patients with NAFLD. METHODS: We searched PubMed, Embase, Cochrane, Scopus, and Web of Science for randomized controlled trials (RCTs) regarding synbiotics supplementation in patients with NAFLD. RESULTS: The meta-analysis revealed that synbiotics supplementation significantly improved liver enzymes (AST, WMD: -9.12 IU/L; 95 % CI: -13.19 to -5.05; ALT, WMD: -8.53 IU/L; 95 % CI: -15.07 to -1.99; GGT, WMD: -10.42 IU/L; 95 % CI: -15.19 to -5.65), lipid profile (TC, WMD: -7.74 mg/dL; 95 % CI: -12.56 to -2.92), obesity indices (body weight, WMD: -1.95 kg; 95 % CI: -3.69 to -0.22; WC, WMD: -1.40 cm; 95 % CI: -2.71 to -0.10), systolic blood pressure (SBP, WMD: -6.00 mmHg; 95 % CI: -11.52 to -0.49), and inflammatory markers (CRP, WMD: -0.69 mg/L; 95 % CI: -1.17 to -0.21; TNF-α, WMD: -14.01 pg/mL; 95 % CI: -21.81 to -6.20). CONCLUSION: Overall, supplementation with synbiotics positively improved liver enzymes, obesity indices, and inflammatory cytokines in patients with NAFLD.


Asunto(s)
Presión Sanguínea , Inflamación , Lípidos , Hígado , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Simbióticos , Humanos , Presión Sanguínea/efectos de los fármacos , Inflamación/dietoterapia , Lípidos/sangre , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Obesidad/dietoterapia , Ensayos Clínicos Controlados Aleatorios como Asunto , Simbióticos/administración & dosificación
11.
Food Res Int ; 195: 114976, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39277213

RESUMEN

Changes in dietary patterns and living habits have led to an increasing number of individuals with elevated cholesterol levels. Excessive consumption of high-cholesterol foods can disrupt the body's lipid metabolism. Numerous studies have firmly established the cholesterol-lowering effects of probiotics and prebiotics, with evidence showing that the synergistic use of synbiotics is functionally more potent than using probiotics or prebiotics alone. Currently, the screening strategy involves screening prebiotics for synbiotic development with probiotics as the core. However, in comparison to probiotics, there are fewer types of prebiotics available, leading to limited resources. Consequently, the combinations of synbiotics obtained are restricted, and probiotics and prebiotics are only relatively suitable. Therefore, in this study, a novel synbiotic screening strategy with prebiotics as the core was developed. The synbiotic combination of Lactobacillus rhamnosus S_82 and xylo-oligosaccharides was screened from the intestinal tract of young people through five generations of xylo-oligosaccharides. Subsequently, the cholesterol-lowering ability of the medium was simulated, and the two carbon sources of glucose and xylo-oligosaccharides were screened out. The results showed that synbiotics may participate in cholesterol-lowering regulation by down-regulating the expression of NPC1L1 gene, down-regulating ACAT2 and increasing the expression of ABCG8 gene in vitro through cell adsorption and cell absorption in vitro, and regulating the intestinal microbiota. Synbiotics hold promise as potential candidates for the prevention of hypercholesterolemia in humans and animals, and this study providing a theoretical foundation for the development of new synbiotic products.


Asunto(s)
Lacticaseibacillus rhamnosus , Oligosacáridos , Prebióticos , Simbióticos , Lacticaseibacillus rhamnosus/metabolismo , Oligosacáridos/farmacología , Humanos , Hipolipemiantes/farmacología , Colesterol/metabolismo , Colesterol/sangre , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos , Glucuronatos
12.
J Affect Disord ; 368: 547-554, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39299595

RESUMEN

BACKGROUND: A growing body of studies revealed that enteric dysbacteriosis could result in depression via the "gut-microbiota-brain axis" (GMBA). Whether probiotics, prebiotics, and synbiotics supplements could lessen the risk of depression is a topic attracting attention. This research was conducted to evaluate the relationship between probiotics, prebiotics, synbiotics, or yogurt supplements and depression with large cross-sectional data. METHODS: All data in our research was sourced from the National Health and Nutrition Examination Survey (NHANES) (2005-2016). Probiotics, prebiotics, synbiotics, and yogurt supplements were identified using Food Frequency Questionnaire (FFQ) and Dietary Supplement Use 30-Day (DSQ). We employed the Patient Health Questionnaire (PHQ-9) for evaluating depression. Logistic regression and the Kaplan-Meier curve were performed to examine the correlation between the supplements and depression, as well as mortality. RESULTS: A total of 17,745 adult participants were selected. The participants who supplemented probiotics, prebiotics, synbiotics, or yogurt products in the last 30 days showed a significantly lower depression rate compared with those who didn't. Specifically, the supplements could alleviate depressive symptoms including sad, anhedonia, sleep problems, fatigue, appetite changes, and psychomotor changes. This association was more prominent in specific populations such as the population aged 40-60 years, male, whites. The supplements also show more significant effects on increasing survival rates in patients with mild depression. LIMITATION: Cross-sectional analysis reveals correlative but not causative association. CONCLUSION: Based on the analysis of NHANES data, our research highlights the positive effect the supplements have on preventing depression, relieving depressive symptoms and increasing survival rates. This effect varied across populations.

13.
Sci Rep ; 14(1): 21372, 2024 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-39266591

RESUMEN

This study was designed to investigate the effect of vitamin D and/or synbiotics on the response to treatment, cytokines profile and hormonal biomarkers in breast cancer patients undergoing neoadjuvant therapy. A total of 76 patients were recruited and completed the course of the intervention between 2019 and 2021 in Kerman, Iran. breast cancer patients were randomly enrolled in this study. Patients divided into four groups to receive one of the following regimens: placebo, vitamin D, synbiotics and a combination of vitamin D and synbiotics. clinicopathologic parameters, inflammatory and anti-inflammatory biomarkers and hormonal levels were measured at the baseline and four months after intervention. The study results found no clear link between the interventions and achieving pathological complete response (pCR), and a similar trend was observed in Ki-67 index examination. After neoadjuvant therapy, TNF-α concentrations decreased, with vitamin D supplementation moderating this decline. Vitamin D supplemented groups showed a significant increase in serum IL-6 levels. While IL-10 levels decreased in the placebo group, all intervention groups were protected from this decline. Moreover, there was a notable increase in the anti-inflammatory index, particularly in the group receiving both vitamin D and synbiotic supplementation, suggesting potential synergistic anti-inflammatory effects from their combined administration. The outcomes suggest a potential anti-inflammatory function of this combination. Consequently, more extensive studies with prolonged follow-up periods and substantial sample sizes are warranted to thoroughly evaluate their potential benefits for breast cancer patients.


Asunto(s)
Neoplasias de la Mama , Citocinas , Simbióticos , Vitamina D , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Vitamina D/sangre , Vitamina D/administración & dosificación , Simbióticos/administración & dosificación , Persona de Mediana Edad , Proyectos Piloto , Citocinas/sangre , Citocinas/metabolismo , Adulto , Terapia Neoadyuvante/métodos , Irán , Resultado del Tratamiento , Sinergismo Farmacológico , Suplementos Dietéticos
14.
Clin Nutr ESPEN ; 63: 813-820, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39178987

RESUMEN

The intricate ecosystem of microorganisms residing within and on the human body, collectively known as the microbiome, significantly influences human health. Imbalances in this microbiome, referred to as dysbiosis, have been associated with various diseases, prompting the exploration of novel therapeutic approaches. Personalized medicine, Tailors treatments to individual patient characteristics, offers a promising avenue for addressing microbiome-related health issues. This review highlights recent developments in utilizing personalized medicine to target the microbiome, aiming to enhance health outcomes. Noteworthy strategies include fecal microbiota transplantation (FMT), where healthy donor microbes are transferred to patients, showing promise in treating conditions such as recurrent Clostridium difficile infection. Additionally, probiotics, which are live microorganisms similar to beneficial gut inhabitants, and prebiotics, non-digestible compounds promoting microbial growth, are emerging as tools to restore microbiome balance. The integration of these approaches, known as synbiotics, enhances microbial colonization and therapeutic effects. Advances in metagenomics and sequencing technologies provide the means to understand individual microbiome profiles, enabling tailored interventions. This paper aims to present the latest insights in leveraging personalized medicine to address microbiome-related health concerns, envisioning a future where microbiome-based therapies reshape disease management and promote human health.


Asunto(s)
Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Prebióticos , Medicina de Precisión , Probióticos , Humanos , Probióticos/uso terapéutico , Disbiosis/terapia , Microbiota , Simbióticos
15.
Microorganisms ; 12(8)2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39203424

RESUMEN

Probiotics are "live microorganisms which, when administered in adequate amount, confer health benefits on the host". They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.

16.
Diabetes Metab Syndr ; 18(8): 103110, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39213690

RESUMEN

BACKGROUND: We aimed to explore how probiotics, prebiotics, or synbiotics impact glycemic indices in patients with diabetes mellitus. METHOD: A comprehensive search was conducted on PubMed, Scopus, and Web of Science from inception up to April 2023. The random-effects model was employed for the study analysis. Furthermore, sensitivity and subgroup analyses were conducted to investigate potential sources of heterogeneity. AMSTAR2 checklist was used to determine the quality of studies. Comprehensive meta-analysis version 3 was used for the study analysis. RESULT: A total of 31 studies were included in the final analysis. Based on the results of the meta-analysis, gut microbial therapy could significantly decrease serum fasting blood glucose levels in patients with type 2 diabetes mellitus (effect size: -0.211; 95 % CI: -0.257, -0.164; P < 0.001). Additionally, significant associations were also found between gut microbial therapy and improved serum levels of fasting insulin, glycated hemoglobin, and homeostatic model assessment for insulin resistance (effect size: -0.087; 95 % confidence interval: -0.120, -0.053; P < 0.001; effect size: -0.166; 95 % confidence interval: -0.200, -0.132; P < 0.001; effect size: -0.230; 95 % confidence interval: -0.288, -0.172; P < 0.001, respectively). CONCLUSION: Our results revealed promising effects of gut microbiota modulation on glycemic profile of patients with type 2 diabetes mellitus. The use of these agents as additional treatments can be considered.

17.
J Microbiol Biotechnol ; 34(9): 1739-1747, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39099195

RESUMEN

Alzheimer's disease (AD) is a progressive neurological disorder that represents a major cause of dementia worldwide. Its pathogenesis involves multiple pathways, including the amyloid cascade, tau protein, oxidative stress, and metal ion dysregulation. Recent studies have suggested a critical link between changes in gut microbial diversity and the disruption of the gut-brain axis in AD. Previous studies primarily explored the potential benefits of probiotics and prebiotics in managing AD. However, studies have yet to fully describe a novel promising approach involving the use of synbiotics, which include a combination of active probiotics and new-generation prebiotics. Synbiotics show potential for mitigating the onset and progression of AD, thereby offering a holistic approach to address the multifaceted nature of AD. This review article primarily aims to gain further insights into the mechanisms of AD, specifically the intricate interaction between gut bacteria and the brain via the gut-brain axis. By understanding this relationship, we can identify potential targets for intervention and therapeutic strategies to combat AD effectively. This review also discusses substantial evidence supporting the role of synbiotics as a promising AD treatment that surpasses traditional probiotic or prebiotic interventions. We find that synbiotics may be used not only to address cognitive decline but also to reduce AD-related psychological burden, thus enhancing the overall quality of life of patients with AD.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Probióticos , Simbióticos , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/prevención & control , Simbióticos/administración & dosificación , Humanos , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Salud Mental , Eje Cerebro-Intestino/fisiología , Prebióticos , Animales , Encéfalo , Calidad de Vida
18.
Surg Today ; 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39158605

RESUMEN

PURPOSE: Infectious complications, particularly post-transplant sepsis, have a critical impact on postoperative outcomes. This study examined the effects of perioperative synbiotic treatment on postoperative outcomes in patients receiving early enteral nutrition. METHODS: We reviewed 210 living-donor liver transplantation procedures and retrospectively analyzed the postoperative outcomes with and without perioperative synbiotic treatment (live lactic acid bacteria, bifidobacteria, and oligosaccharides) 5 days before and after living-donor liver transplantation. RESULTS: The synbiotic group (n = 34) had significantly fewer male donors (38.2% vs. 61.9%, p = 0.011) and a higher proportion of ABO-incompatible grafts (52.9% vs. 25.6%, p = 0.021) than the non-synbiotic group (n = 176). The incidence of sepsis was significantly lower in the synbiotic group than in the non-synbiotic group (0% vs. 7.4%, p = 0.029), with a lower incidence rate of sepsis due to bacteremia with intestinal bacteria (0% vs. 4.6%, p = 0.089). There were no significant differences in the proportions of acute rejection, small-for-size graft syndrome, or postoperative liver function between the two groups. Furthermore, there was no significant difference in the graft survival rates after LDLT between two groups. (p = 0.24). CONCLUSION: Perioperative synbiotic treatment prevents post-transplant sepsis, even with early enteral nutrition.

19.
Cureus ; 16(8): e66994, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39161553

RESUMEN

BACKGROUND AND AIMS: The use of gut biotics, including probiotics, prebiotics, and synbiotics, has shown substantial potential in the management of various health conditions possibly through the gut-organ axis. The role of gut biotics in modulating the gut-brain axis is becoming evident with more research focusing on this intervention. Improvement of gut-organ axis function is possible by using food-related products called gut biotics. However, there is limited comprehension of the knowledge and use of these intestinal or gut biotics. Our aim was to recognize knowledge gaps and assess the improvement of understanding following an education intervention. METHODS: A single-arm study encompassing a convenient sample of 161 inpatient and outpatient subjects aged 50 years and older was conducted at the University of Alberta Hospital from June to August 2023. Knowledge about gut biotics was evaluated using a structured questionnaire consisting of 16 questions and involving six thematic areas. To ensure validity, the questionnaire was pre-tested on 10 physicians and residents who were not part of the study. The questionnaire was administered to study subjects prior to receiving an information sheet about gut biotics. Two weeks after receiving the information sheet, all participants were contacted by phone, and the same questionnaire was administered again. Of the 161 patients, 122 completed the pre-intervention and post-intervention questionnaires and were considered in the analysis. RESULTS: The mean age of the participants was 72 years (SD: 10.8), 57% comprised women, and 39% had less than a high school education. The proportion of polypharmacy and multimorbidity was 87% and 97%, respectively. Following the intervention, there was a noticeable enhancement in knowledge across all the themes, with statistical significance (p<0.001) observed in 14 out of 16 questions as determined by the homogeneity statistical test. CONCLUSIONS: Knowledge gaps in gut biotics were prevalent among study participants, and the educational intervention effectively contributed to the enhancement of knowledge. The results of this study provide valuable information for the development of targeted health education strategies focusing on gut biotics, which may play a role in improving gut-organ axis function.

20.
Food Sci Biotechnol ; 33(9): 2065-2080, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39130661

RESUMEN

The human gut microbiome accompanies us from birth, and it is developed and matured by diet, lifestyle, and environmental factors. During aging, the bacterial composition evolves in reciprocal communication with the host's physiological properties. Many diseases are closely related to the gut microbiome, which means the modulation of the gut microbiome can promote the disease targeting remote organs. This review explores the intricate interaction between the gut microbiome and other organs, and their improvement from disease by prebiotics, probiotics, synbiotics, and postbiotics. Each section of the review is supported by clinical trials that substantiate the benefits of modulation the gut microbiome through dietary intervention for improving primary health outcomes across various axes with the gut. In conclusion, the review underscores the significant potential of targeting the gut microbiome for therapeutic and preventative interventions in a wide range of diseases, calling for further research to fully unlock the microbiome's capabilities in enhancing human health.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...