Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 771
Filter
1.
J Drugs Dermatol ; 23(7): 557-563, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38954628

ABSTRACT

Variations in the epidemiology, clinical presentation, and disease course in atopic dermatitis (AD) patients with Skin of Color (SOC) compared with white counterparts have been reported. In this study, we evaluated the capability of a new imaging device (SkinCam) in quantifying skin texture changes in diverse patients, presenting with AD or xerosis, after using a prebiotic skincare routine over 10 weeks.  A total of 39 subjects from diverse racial/ethnic backgrounds, aged 3 to 76 years old, with Fitzpatrick skin phototypes I to VI, presenting with mild AD and moderate to severe xerosis, were enrolled in the study. All subjects used a prebiotic cleanser on its own for 2 weeks, followed by a prebiotic moisturizer in conjunction for an additional 8 weeks. Standardized images of the subjects' legs were taken with SkinCam at several time points (baseline, week 2, and week 10), and analyzed for skin texture parameters. Our results demonstrate that both skin texture irregularity and skin color patterns significantly improve over time with a prebiotic skincare regimen in AD (n=12) and xerosis (n=24) subjects. Interestingly, image analyses showed more improvement over time in xerosis and AD SOC patients (n=18, Fitzpatrick IV-VI). Lastly, skin texture analyses from SkinCam imaging correlated with clinical assessments, showing significant improvement by prebiotic skincare regimen in all subjects by week 10. In summary, our results demonstrate that the SkinCam imaging device has the capability to effectively monitor skin texture parameters over time in both AD and xerosis patients with lightly and darkly pigmented skin. J Drugs Dermatol. 2024;23(7):557-563.  doi:10.36849/JDD.8371.


Subject(s)
Dermatitis, Atopic , Prebiotics , Skin Care , Skin Pigmentation , Humans , Dermatitis, Atopic/diagnosis , Adult , Middle Aged , Aged , Female , Prebiotics/administration & dosage , Male , Young Adult , Adolescent , Skin Pigmentation/drug effects , Skin Care/methods , Child , Child, Preschool , Ethnicity/statistics & numerical data , Treatment Outcome , Skin Cream/administration & dosage
2.
Front Immunol ; 15: 1410928, 2024.
Article in English | MEDLINE | ID: mdl-38903520

ABSTRACT

In recent years, with the deepening understanding of the gut microbiota, it has been recognized to play a significant role in the development and progression of diseases. Particularly in gastrointestinal tumors, the gut microbiota influences tumor growth by dysbiosis, release of bacterial toxins, and modulation of host signaling pathways and immune status. Immune checkpoint inhibitors (ICIs) have greatly improved cancer treatment efficacy by enhancing immune cell responses. Current clinical and preclinical studies have demonstrated that the gut microbiota and its metabolites can enhance the effectiveness of immunotherapy. Furthermore, certain gut microbiota can serve as biomarkers for predicting immunotherapy responses. Interventions targeting the gut microbiota for the treatment of gastrointestinal diseases, especially colorectal cancer (CRC), include fecal microbiota transplantation, probiotics, prebiotics, engineered bacteria, and dietary interventions. These approaches not only improve the efficacy of ICIs but also hold promise for enhancing immunotherapy outcomes. In this review, we primarily discuss the role of the gut microbiota and its metabolites in tumors, host immunity, and immunotherapy.


Subject(s)
Gastrointestinal Microbiome , Immunotherapy , Humans , Gastrointestinal Microbiome/immunology , Immunotherapy/methods , Animals , Dysbiosis/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/microbiology , Probiotics/therapeutic use , Fecal Microbiota Transplantation , Immune Checkpoint Inhibitors/therapeutic use , Prebiotics/administration & dosage
3.
Adv Genet ; 111: 237-310, 2024.
Article in English | MEDLINE | ID: mdl-38908901

ABSTRACT

Microorganisms have been used in nutrition and medicine for thousands of years worldwide, long before humanity knew of their existence. It is now known that the gut microbiota plays a key role in regulating inflammatory, metabolic, immune and neurobiological processes. This text discusses the importance of microbiota-based precision nutrition in gut permeability, as well as the main advances and current limitations of traditional probiotics, new-generation probiotics, psychobiotic probiotics with an effect on emotional health, probiotic foods, prebiotics, and postbiotics such as short-chain fatty acids, neurotransmitters and vitamins. The aim is to provide a theoretical context built on current scientific evidence for the practical application of microbiota-based precision nutrition in specific health fields and in improving health, quality of life and physiological performance.


Subject(s)
Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Probiotics/administration & dosage , Prebiotics/administration & dosage , Precision Medicine/methods
4.
Microbiome ; 12(1): 113, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907315

ABSTRACT

BACKGROUND: Although the microbiota has been extensively associated with HIV pathogenesis, the majority of studies, particularly those using omics techniques, are largely correlative and serve primarily as a basis for hypothesis generation. Furthermore, most have focused on characterizing the taxonomic composition of the bacterial component, often overlooking other levels of the microbiome. The intricate mechanisms by which the microbiota influences immune responses to HIV are still poorly understood. Interventional studies on gut microbiota provide a powerful tool to test the hypothesis of whether we can harness the microbiota to improve health outcomes in people with HIV. RESULTS: Here, we review the multifaceted role of the gut microbiome in HIV/SIV disease progression and its potential as a therapeutic target. We explore the complex interplay between gut microbial dysbiosis and systemic inflammation, highlighting the potential for microbiome-based therapeutics to open new avenues in HIV management. These include exploring the efficacy of probiotics, prebiotics, fecal microbiota transplantation, and targeted dietary modifications. We also address the challenges inherent in this research area, such as the difficulty in inducing long-lasting microbiome alterations and the complexities of study designs, including variations in probiotic strains, donor selection for FMT, antibiotic conditioning regimens, and the hurdles in translating findings into clinical practice. Finally, we speculate on future directions for this rapidly evolving field, emphasizing the need for a more granular understanding of microbiome-immune interactions, the development of personalized microbiome-based therapies, and the application of novel technologies to identify potential therapeutic agents. CONCLUSIONS: Our review underscores the importance of the gut microbiome in HIV/SIV disease and its potential as a target for innovative therapeutic strategies.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , HIV Infections , Probiotics , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Dysbiosis/therapy , Dysbiosis/microbiology , Humans , HIV Infections/microbiology , HIV Infections/therapy , HIV Infections/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/microbiology , Probiotics/therapeutic use , Animals , Prebiotics/administration & dosage , HIV/physiology
6.
Nutrients ; 16(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38892505

ABSTRACT

Several studies show that gut microbiotas in patients with nonalcoholic fatty liver disease (NAFLD) differ from those in a healthy population, suggesting that this alteration plays a role in NAFLD pathogenesis. We investigated whether prebiotic administration affects liver fat content and/or liver-related and metabolic parameters. Patients with NAFLD and metabolic syndrome (age: 50 ± 11; 79% men) were randomized to receive either 16 g/day of prebiotic (ITFs-inulin-type fructans) (n = 8) or placebo (maltodextrin) (n = 11) for 12 weeks. Patients were instructed to maintain a stable weight throughout the study. Liver fat content (measured by H1MRS), fecal microbiota, and metabolic, inflammatory, and liver parameters were determined before and after intervention. Fecal samples from patients who received the prebiotic had an increased content of Bifidobacterium (p = 0.025), which was not observed with the placebo. However, the baseline and end-of-study liver fat contents did not change significantly in the prebiotic and placebo groups, neither did the liver function tests' metabolic and inflammatory mediators, including fibroblast growth factor-19 and lipopolysaccharide-binding protein. Body weight remained stable in both groups. These findings suggest that prebiotic treatment without weight reduction is insufficient to improve NAFLD.


Subject(s)
Feces , Gastrointestinal Microbiome , Liver , Non-alcoholic Fatty Liver Disease , Prebiotics , Humans , Non-alcoholic Fatty Liver Disease/diet therapy , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/microbiology , Prebiotics/administration & dosage , Male , Middle Aged , Female , Pilot Projects , Adult , Liver/metabolism , Feces/microbiology , Bifidobacterium , Double-Blind Method , Metabolic Syndrome/diet therapy , Metabolic Syndrome/therapy
7.
Nutrients ; 16(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38892536

ABSTRACT

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.


Subject(s)
Athletes , Athletic Performance , Exercise , Gastrointestinal Microbiome , Probiotics , Humans , Athletic Performance/physiology , Probiotics/administration & dosage , Gastrointestinal Microbiome/physiology , Exercise/physiology , Prebiotics/administration & dosage , Synbiotics/administration & dosage , Energy Metabolism , Oxidative Stress , Dietary Supplements , Post-Exercise Recovery
8.
Nutrients ; 16(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892608

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Glycated Hemoglobin , Probiotics , Randomized Controlled Trials as Topic , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/blood , Humans , Gastrointestinal Microbiome/drug effects , Glycated Hemoglobin/metabolism , Probiotics/therapeutic use , Prebiotics/administration & dosage , Biomarkers/blood , Synbiotics/administration & dosage , Dietary Supplements , Female , Dysbiosis , Adult , Male
9.
Life Sci ; 350: 122748, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38843992

ABSTRACT

Neurodegenerative diseases (NDs) are a group of heterogeneous disorders with a high socioeconomic burden. Although pharmacotherapy is currently the principal therapeutic approach for the management of NDs, mounting evidence supports the notion that the protracted application of available drugs would abate their dopaminergic outcomes in the long run. The therapeutic application of microbiome-based modalities has received escalating attention in biomedical works. In-depth investigations of the bidirectional communication between the microbiome in the gut and the brain offer a multitude of targets for the treatment of NDs or maximizing the patient's quality of life. Probiotic administration is a well-known microbial-oriented approach to modulate the gut microbiota and potentially influence the process of neurodegeneration. Of note, there is a strong need for further investigation to map out the mechanistic prospects for the gut-brain axis and the clinical efficacy of probiotics. In this review, we discuss the importance of microbiome modulation and hemostasis via probiotics, prebiotics, postbiotics and synbiotics in ameliorating pathological neurodegenerative events. Also, we meticulously describe the underlying mechanism of action of probiotics and their metabolites on the gut-brain axis in different NDs. We suppose that the present work will provide a functional direction for the use of probiotic-based modalities in promoting current practical treatments for the management of neurodegenerative-related diseases.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Neurodegenerative Diseases , Probiotics , Probiotics/therapeutic use , Humans , Gastrointestinal Microbiome/physiology , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/therapy , Brain-Gut Axis/physiology , Animals , Brain/metabolism , Prebiotics/administration & dosage
10.
Clin Nutr ; 43(7): 1832-1849, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878554

ABSTRACT

BACKGROUND AND AIMS: Microbiota plays an essential role in maintaining body health, through positive influences on metabolic, defensive, and trophic processes and on intercellular communication. Imbalance in intestinal flora, with the proliferation of harmful bacterial species (dysbiosis) is consistently reported in chronic illnesses, including neurodegenerative diseases (ND). Correcting dysbiosis can have a beneficial impact on the symptoms and evolution of ND. This review examines the effects of microbiota modulation through administration of probiotics, prebiotics, symbiotics, or prebiotics' metabolites (postbiotics) in patients with ND like multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS). METHODS: PubMed, Web of Science, Medline databases and ClinicalTrials.gov registry searches were performed using pre-/pro-/postbiotics and ND-related terms. Further references were obtained by checking relevant articles. RESULTS: Although few compared to animal studies, the human studies generally show positive effects on disease-specific symptoms, overall health, metabolic parameters, on oxidative stress and immunological markers. Therapy with probiotics in various forms (mixtures of bacterial strains, fecal microbiota transplant, diets rich in fermented foods) exert favorable effects on patients' mental health, cognition, and quality of life, targeting pathogenetic ND mechanisms and inducing reparatory mechanisms at the cellular level. More encouraging results have been observed in prebiotic/postbiotic therapy in some ND. CONCLUSIONS: The effects of probiotic-related interventions depend on the patients' ND stage and pre-existing allopathic medication. Further studies on larger cohorts and long term comprehensive neuropsychiatric, metabolic, biochemical testing, and neuroimaging monitoring are necessary to optimize therapeutic protocols in ND.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Neurodegenerative Diseases , Prebiotics , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/therapy , Probiotics/administration & dosage , Probiotics/therapeutic use , Prebiotics/administration & dosage , Dysbiosis/therapy , Dysbiosis/microbiology , Animals , Fecal Microbiota Transplantation
11.
Int J Mol Sci ; 25(11)2024 May 26.
Article in English | MEDLINE | ID: mdl-38891970

ABSTRACT

Nowadays, depressive disorder is spreading rapidly all over the world. Therefore, attention to the studies of the pathogenesis of the disease in order to find novel ways of early diagnosis and treatment is increasing among the scientific and medical communities. Special attention is drawn to a biomarker and therapeutic strategy through the microbiota-gut-brain axis. It is known that the symbiotic interactions between the gut microbes and the host can affect mental health. The review analyzes the mechanisms and ways of action of the gut microbiota on the pathophysiology of depression. The possibility of using knowledge about the taxonomic composition and metabolic profile of the microbiota of patients with depression to select gene compositions (metagenomic signature) as biomarkers of the disease is evaluated. The use of in silico technologies (machine learning) for the diagnosis of depression based on the biomarkers of the gut microbiota is given. Alternative approaches to the treatment of depression are being considered by balancing the microbial composition through dietary modifications and the use of additives, namely probiotics, postbiotics (including vesicles) and prebiotics as psychobiotics, and fecal transplantation. The bacterium Faecalibacterium prausnitzii is under consideration as a promising new-generation probiotic and auxiliary diagnostic biomarker of depression. The analysis conducted in this review may be useful for clinical practice and pharmacology.


Subject(s)
Depression , Gastrointestinal Microbiome , Probiotics , Humans , Depression/therapy , Depression/microbiology , Depression/diagnosis , Probiotics/therapeutic use , Biomarkers , Fecal Microbiota Transplantation , Brain-Gut Axis , Prebiotics/administration & dosage
12.
Sci Rep ; 14(1): 12838, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834634

ABSTRACT

Disorders related to gut health are a significant cause of morbidity among athletes in wheelchair. This pilot feasibility trial aims to investigate whether probiotics compared to prebiotics can improve inflammatory status and gut microbiome composition in elite athletes in wheelchair. We conducted a 12-week, randomized, cross-over controlled trial involving 14 elite Swiss athletes in wheelchair. Participants were given a multispecies-multistrain probiotic or prebiotic (oat bran) daily for 4 weeks (Clinical trials.gov NCT04659408 09/12/2020). This was followed by a 4-week washout and then crossed over. Thirty inflammatory markers were assessed using bead-based multiplex immunoassays (LegendPlex) from serum samples. The gut microbiome was characterized via 16S rRNA sequencing of stool DNA samples. Statistical analyses were conducted using linear mixed-effect models (LMM). At baseline, most athletes (10/14) exhibited low levels of inflammation which associated with higher gut microbiome alpha diversity indices compared to those with high inflammation levels. The use of probiotic had higher decrease in 25 (83%) inflammatory markers measured compared to prebiotic use. Probiotic has the potential in lowering inflammation status and improving the gut microbiome diversity. The future trial should focus on having sufficient sample sizes, population with higher inflammation status, longer intervention exposure and use of differential abundance analysis.


Subject(s)
Athletes , Cross-Over Studies , Gastrointestinal Microbiome , Inflammation , Prebiotics , Probiotics , Humans , Probiotics/administration & dosage , Probiotics/therapeutic use , Prebiotics/administration & dosage , Male , Pilot Projects , Adult , Female , Wheelchairs , Young Adult , RNA, Ribosomal, 16S/genetics , Biomarkers , Feces/microbiology
13.
Nutrients ; 16(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38931292

ABSTRACT

Type 2 diabetes is a disease with significant health consequences for the individual. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. One of them is the association of type 2 diabetes with microbiota. Through the enteric nervous system and the gut-microbiota axis, the microbiota affects the functioning of the body. It has been proven to have a real impact on influencing glucose and lipid metabolism and insulin sensitivity. With dysbiosis, there is increased bacterial translocation through the disrupted intestinal barrier and increased inflammation in the body. In diabetes, the microbiota's composition is altered with, for example, a more abundant class of Betaproteobacteria. The consequences of these disorders are linked to mechanisms involving short-chain fatty acids, branched-chain amino acids, and bacterial lipopolysaccharide, among others. Interventions focusing on the gut microbiota are gaining traction as a promising approach to diabetes management. Studies are currently being conducted on the effects of the supply of probiotics and prebiotics, as well as fecal microbiota transplantation, on the course of diabetes. Further research will allow us to fully develop our knowledge on the subject and possibly best treat and prevent type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/therapy , Probiotics/therapeutic use , Prebiotics/administration & dosage , Dysbiosis/therapy , Animals
14.
J Food Drug Anal ; 32(2): 213-226, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38934690

ABSTRACT

Citrus peels contain abundant polyphenols, particularly flavonoids, and have been shown to exert lipid accumulation decreasing ability. In this study, Citrus depressa peel applied to oven drying and extracted with ethanol extract as CDEE to analyze its flavonoids compositions and investigated its effects on a high-fat diet (HFD)-induced obese mice model. CDEE contained several flavonoids such as hesperidin, sinesentin, nobiletin, tangeretin, 5-demethylnobiletin, and 5-demethyltangeretin. The mice fed an HFD, and administration of 2% CDEE to could decrease weight gain, abdominal fat weight, inguinal fat weight, and the adipocyte size, and CDEE also reduced serum total cholesterol (TCHO), triacylglycerol (TG) compared with mice fed only on HFD. CDEE hindered lipid accumulation through a decreased fatty acid synthase (FAS) protein expression via upregulation of the protein expression of AMP-activated protein kinase α (AMPKα). Moreover, CDEE modulated gut microbiota that altered by HFD through an increased abundance of Lactobacillus reuteri compared with the HFD group. The results demonstrated that CDEE helps decrease lipid accumulation through the AMPK pathway, which also indicates a prebiotic-like effect on gut microbiota.


Subject(s)
Citrus , Diet, High-Fat , Gastrointestinal Microbiome , Lipid Metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity , Plant Extracts , Prebiotics , Animals , Gastrointestinal Microbiome/drug effects , Mice , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Citrus/chemistry , Male , Obesity/metabolism , Obesity/drug therapy , Lipid Metabolism/drug effects , Prebiotics/administration & dosage , Prebiotics/analysis , Diet, High-Fat/adverse effects , Humans , Triglycerides/metabolism , Triglycerides/blood , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , Bacteria/drug effects
15.
JPEN J Parenter Enteral Nutr ; 48(5): 538-545, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38689534

ABSTRACT

Focused perioperative nutrition strategies have proven benefits on the outcomes for patients undergoing major abdominal surgery. In this brief article, we will review these strategies and the evidence to support them with a focus on gastrointestinal anastomotic healing. We will elaborate the risks and benefits of enteral feeds, immune- and metabolic-modulating formulas, prebiotics and probiotics, and prehabilitation in preparation for surgery. Additionally, we will discuss the role of fish oils (eicosapentaenoic acid and docosahexaenoic acid) in the surgical patient and new data on specialized proresolving mediators in inflammation resolution. Finally, this article will consider the harmful impact surgical trauma has on the microbiome and the potential for perioperative dietary modulation to attenuate these negative effects.


Subject(s)
Enteral Nutrition , Perioperative Care , Prebiotics , Probiotics , Humans , Perioperative Care/methods , Prebiotics/administration & dosage , Enteral Nutrition/methods , Probiotics/administration & dosage , Gastrointestinal Microbiome , Fish Oils/administration & dosage , Digestive System Surgical Procedures/methods , Postoperative Complications/prevention & control , Wound Healing , Inflammation
16.
Am J Physiol Endocrinol Metab ; 327(1): E89-E102, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38809510

ABSTRACT

The gut microbiome, a complex assembly of microorganisms, significantly impacts human health by influencing nutrient absorption, the immune system, and disease response. These microorganisms form a dynamic ecosystem that is critical to maintaining overall well-being. Prebiotics and probiotics are pivotal in regulating gut microbiota composition. Prebiotics nourish beneficial bacteria and promote their growth, whereas probiotics help maintain balance within the microbiome. This intricate balance extends to several aspects of health, including maintaining the integrity of the gut barrier, regulating immune responses, and producing metabolites crucial for metabolic health. Dysbiosis, or an imbalance in the gut microbiota, has been linked to metabolic disorders such as type 2 diabetes, obesity, and cardiovascular disease. Impaired gut barrier function, endotoxemia, and low-grade inflammation are associated with toll-like receptors influencing proinflammatory pathways. Short-chain fatty acids derived from microbial fermentation modulate anti-inflammatory and immune system pathways. Prebiotics positively influence gut microbiota, whereas probiotics, especially Lactobacillus and Bifidobacterium strains, may improve metabolic outcomes, such as glycemic control in diabetes. It is important to consider strain-specific effects and study variability when interpreting these findings, highlighting the need for further research to optimize their therapeutic potential. The aim of this report is therefore to review the role of the gut microbiota in metabolic health and disease and the effects of prebiotics and probiotics on the gut microbiome and their therapeutic role, integrating a broad understanding of physiological mechanisms with a clinical perspective.


Subject(s)
Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Prebiotics/administration & dosage , Animals , Dysbiosis/microbiology , Metabolic Diseases/microbiology , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/immunology
17.
Adv Drug Deliv Rev ; 210: 115332, 2024 07.
Article in English | MEDLINE | ID: mdl-38759702

ABSTRACT

Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.


Subject(s)
Antineoplastic Agents , Gastrointestinal Microbiome , Neoplasms , Prebiotics , Probiotics , Humans , Neoplasms/drug therapy , Neoplasms/therapy , Probiotics/administration & dosage , Prebiotics/administration & dosage , Antineoplastic Agents/administration & dosage , Animals , Immunotherapy/methods
18.
Clin Nutr ; 43(6): 1433-1446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704983

ABSTRACT

The prevalence of childhood and adolescent obesity has globally reached alarming dimensions and many adolescents affected by obesity already present one or more obesity-related comorbidities. In recent years, emerging evidence supporting the role of gut microbiota in the pathophysiology of metabolic diseases has been reported and the use of prebiotics, probiotics, synbiotics and postbiotics as a strategy to manipulate gut microbiota has become popular. The aim of this review is to explore the relationship between gut microbiota and metabolic syndrome in adolescents and to discuss the potential use of prebiotics, probiotics, synbiotics and postbiotics for the prevention and treatment of this clinical picture in adolescence. According to the most recent literature, prebiotics, probiotics and synbiotics have no clear effect on MetS, but a possible modulation of anthropometric parameters has been observed after synbiotic supplementation. Only one study has examined the role of postbiotics in alleviating metabolic complications in children with obesity but not in adolescents. More extensive research is needed to support the conclusions drawn so far and to develop effective microbiome-based interventions that may help improving the quality of life of children and adolescents exposed to the increasing prevalence of MetS.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome , Pediatric Obesity , Prebiotics , Probiotics , Synbiotics , Humans , Metabolic Syndrome/therapy , Metabolic Syndrome/microbiology , Prebiotics/administration & dosage , Probiotics/administration & dosage , Probiotics/therapeutic use , Synbiotics/administration & dosage , Adolescent , Pediatric Obesity/therapy , Pediatric Obesity/microbiology , Child
19.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-38783711

ABSTRACT

Dietary fibers and biotics have been shown to support gastrointestinal health in dogs, but are usually tested individually. There is value in testing fiber-biotic combinations that are commonly used commercially. Therefore, this study was conducted to determine the apparent total tract macronutrient digestibility (ATTD) of diets supplemented with fibers or biotics and to evaluate their effects on the fecal characteristics, metabolites, microbiota, and immunoglobulin A (IgA) concentrations of dogs. Twelve healthy adult female beagle dogs (age = 6.2 ±â€…1.6 yr; body weight = 9.5 ±â€…1.1 kg) were used in a replicated 3 × 3 Latin square design to test three treatments: 1) control diet based on rice, chicken meal, tapioca starch, and cellulose + a placebo treat (CT); 2) diet based on rice, chicken meal, garbanzo beans, and cellulose + a placebo treat (GB); 3) diet based on rice, chicken meal, garbanzo beans, and a functional fiber/prebiotic blend + a probiotic-containing treat (GBPP). In each 28-d period, a 22-d diet adaptation was followed by a 5-d fecal collection phase. Fasted blood samples were collected on day 28. Data were analyzed using the Mixed Models procedure of SAS 9.4, with P < 0.05 being significant and P < 0.10 being trends. ATTD of dry matter (DM), organic matter, and energy were lower (P < 0.001) and DM fecal output was higher (P < 0.01) in dogs fed GBPP than CT or GB, whereas ATTD of crude protein was higher (P < 0.001) in dogs fed CT and GBPP than GB. ATTD of fat was higher (P < 0.001) and wet fecal output was lower (P < 0.01) in dogs fed CT than GB or GBPP. Fecal DM% was higher (P < 0.001) in dogs fed CT than GBPP or GB, and higher in dogs fed GBPP than GB. Fecal short-chain fatty acid concentrations were higher (P < 0.001) in dogs fed GB than CT or GBPP, and higher in dogs fed GB than GBPP. Fecal IgA concentrations were higher (P < 0.01) in dogs fed GB than CT. Fecal microbiota populations were affected by diet, with alpha diversity being higher (P < 0.01) in dogs fed GB than CT, and beta diversity shifting following dietary fiber and biotic supplementation. The relative abundance of 24 bacterial genera was altered in dogs fed GB or GBPP than CT. Serum triglyceride concentrations were lower in dogs fed GB than GBPP or CT. Our results demonstrate that legume-based dietary fibers, with or without prebiotics and probiotics, reduce ATTD, increase stool output, beneficially shift fecal metabolites and microbiota, and reduce blood lipids in adult dogs.


Functional fibers and biotics have demonstrated the potential to modulate the gut microbiome and improve gastrointestinal health in dogs, but are often tested individually. Research investigating unique fiber/biotic combinations is needed. Twelve dogs were used in a replicated 3 × 3 Latin square design to test the efficacy of three dietary treatments on apparent total tract macronutrient digestibility (ATTD) and the fecal characteristics, metabolites, microbiota, and immunoglobulin A concentrations of dogs. Treatments included a low-fiber control diet without prebiotics or probiotics + a placebo treat, a diet containing garbanzo beans + a placebo treat (GB), and a diet containing garbanzo beans and a prebiotic blend + a probiotic (Bacillus subtilis and Bacillus amyloliquefaciens) treat (GBPP). ATTD was reduced and stool output was greater in dogs fed GB or GBPP than controls. Fecal short-chain fatty acids were higher in dogs fed GB or GBPP than controls. Fecal immunoglobulin A was higher, while blood lipids were lower in dogs fed GB than control. Finally, GB and GBPP shifted fecal bacterial populations. Our results demonstrate that legume-based dietary fibers, with or without prebiotics and probiotics, reduce ATTD, increase stool output, beneficially shift fecal metabolites and microbiota, and reduce blood lipids in adult dogs.


Subject(s)
Animal Feed , Diet , Dietary Fiber , Dietary Supplements , Digestion , Feces , Gastrointestinal Microbiome , Animals , Dogs , Dietary Fiber/metabolism , Dietary Fiber/pharmacology , Feces/chemistry , Feces/microbiology , Female , Digestion/drug effects , Digestion/physiology , Animal Feed/analysis , Diet/veterinary , Gastrointestinal Microbiome/drug effects , Animal Nutritional Physiological Phenomena , Nutrients/metabolism , Probiotics/pharmacology , Probiotics/administration & dosage , Prebiotics/administration & dosage , Immunoglobulin A/metabolism
20.
Poult Sci ; 103(7): 103807, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38713991

ABSTRACT

This study aimed to evaluate the effect of low molecular weight Acanthopanax polysaccharides on simulated digestion, probiotics, and intestinal flora of broilers in vitro. The experiments were carried out by H2O2-Vc degradation of Acanthopanax polysaccharides, in vitro simulated digestion to evaluate the digestive performance of polysaccharides with different molecular weights, in vitro probiotic evaluation of the probiotic effect of polysaccharides on lactobacilli and bifidobacteria, in vitro anaerobic fermentation and high-throughput sequencing of 16S rRNA genes to study the impact of Acanthopanax polysaccharides on the intestinal flora of broilers, and the effect of Acanthopanax polysaccharides on the short-chain fatty acids of intestines were determined by GC-MS method. The results showed that the molecular weight of Acanthopanax polysaccharide (ASPS) was 9,543 Da, and the molecular weights of polysaccharides ASPS-1 and ASPS-2 were reduced to 4,288 Da and 3,822 Da after degradation, and the particle sizes, PDIs, and viscosities were also significantly decreased. ASPS-1 has anti-digestive properties and better in vitro probiotic properties. The addition of ASPS-1 regulates the structure of intestinal microorganisms by regulating fecalibacterium to produce short-chain fatty acids, promoting the colonization of beneficial bacteria such as fecalibacterium, paraprevotella and diminishing the prevalence of detrimental bacteria such as Fusobacteria. Interestingly the ASPS-1 group found higher levels of Paraprevotella, which degraded trypsin in the gut, reducing inflammation, acted as a gut protector, and was influential in increasing the levels of acetic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, and total SCFAs in the fermented feces. Therefore, the degraded ASPS-1 can better regulate the structure of intestinal flora and promote the production of SCFAs, creating possibilities for its use as a potential prebiotic, which is conducive to the intestinal health of poultry.


Subject(s)
Chickens , Digestion , Eleutherococcus , Feces , Fermentation , Gastrointestinal Microbiome , Polysaccharides , Prebiotics , Animals , Gastrointestinal Microbiome/drug effects , Chickens/microbiology , Eleutherococcus/chemistry , Polysaccharides/pharmacology , Polysaccharides/metabolism , Prebiotics/administration & dosage , Prebiotics/analysis , Feces/microbiology , Digestion/drug effects , Animal Feed/analysis , Diet/veterinary , Fatty Acids, Volatile/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...