Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.920
Filter
1.
Vet Med Sci ; 10(5): e1550, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39119788

ABSTRACT

BACKGROUND: A prebiotic is defined as an indigestible feed substance that beneficially affects the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the large intestine, thereby improving host health and products. OBJECTIVES: This study was conducted to determine the effects of supplementing prebiotic fructooligosaccharide (FOS) to the diets of Hy-Line W-36 laying hens. METHODS: A total of 168 Hy-Line W-36 laying hens were allocated to four dietary levels of FOS (0, 1.0, 2.0, 3.0 g/kg diet), 6 replicates of 7 birds each during 63-74. The experiment was performed using a completely randomized design. RESULTS: Productive performance was not significantly affected by the FOS supplementation. Body weight gain was linearly decreased with increasing FOS levels in the diet (p < 0.01). However, eggshell strength, shell thickness and Ca and p percentages were not significantly affected, as were anti-sheep red blood cell titres, blood parameters and blood metabolites. In the first period of the experiment (63-65 weeks), shape index and Haugh unit at the dose of 3.0 g/kg FOS were significantly increased and decreased in comparison with control, respectively (p < 0.05). In the third and fourth periods (69-71 and 72-74 weeks of age), the FOS had no significant effect on the internal egg quality traits. Furthermore, FOS had a linear decrease in the most saturated fatty acids (SFAs), including myristic, palmitic, margaric and stearic fatty acids; some of the mono-un-SFA (MUFA; palmitoleic and ginkgolic acids), and poly-unsaturated fatty acids (γ-linolenic and eicosatrienoic). CONCLUSIONS: Supplementing different levels of FOS to the diet of commercial laying hens had no significant effect on the layers' performance, immune response and blood parameters, whereas there was a significant effect on some of the internal egg quality traits and egg yolk fatty acid contents.


Subject(s)
Animal Feed , Chickens , Diet , Dietary Supplements , Oligosaccharides , Animals , Chickens/physiology , Chickens/immunology , Chickens/blood , Oligosaccharides/administration & dosage , Female , Animal Feed/analysis , Diet/veterinary , Dietary Supplements/analysis , Prebiotics/analysis , Dose-Response Relationship, Drug , Random Allocation , Eggs/analysis , Animal Nutritional Physiological Phenomena/drug effects
2.
Pediatr Pulmonol ; 59 Suppl 1: S70-S80, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39105345

ABSTRACT

People with cystic fibrosis (pwCF) have an altered gastrointestinal microbiome. These individuals also demonstrate propensity toward developing small intestinal bacterial overgrowth (SIBO). The dysbiosis present has intestinal and extraintestinal implications, including potential links with the higher rates of gastrointestinal malignancies described in CF. Given these implications, there is growing interest in therapeutic options for microbiome modulation. Alternative therapies, including probiotics and prebiotics, and current CF transmembrane conductance regulator gene modulators are promising interventions for ameliorating gut microbiome dysfunction in pwCF. This article will characterize and discuss the current state of knowledge and expert opinions on gut dysbiosis and SIBO in the context of CF, before reviewing the current evidence supporting gut microbial modulating therapies in CF.


Subject(s)
Cystic Fibrosis , Dysbiosis , Gastrointestinal Microbiome , Intestine, Small , Probiotics , Cystic Fibrosis/microbiology , Humans , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , Dysbiosis/microbiology , Intestine, Small/microbiology , Prebiotics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics
3.
Sci Rep ; 14(1): 18397, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117977

ABSTRACT

Antibiotics, improper food, and stress have created a dysbiotic state in the gut and almost 81% of the world's population has been affected due to the pandemic of COVID-19 and the prevalence of dengue virus in the past few years. The main intent of this study is to synthesize nanosynbiotics as nu traceuticals by combining probiotics, and prebiotics with nanoformulation. The effectiveness of the nanosynbiotics was evaluated using a variety of Nutra-pharmacogenetic assays leading to an AI-integrated formulation profiling was assessed by using machine learning methods. Consequently, Acetobacter oryzoeni as a probiotic and inulin as a prebiotic has been chosen and iron-mediated nanoformulation of symbiotic is achieved. Nanosynbiotics possessed 89.4, 96.7, 93.57, 83.53, 88.53% potential powers of Nutra-pharmacogenetic assays. Artificial intelligent solid dispersion formulation of nanosynbiotics has high dissolution, absorption, distribution, and synergism, in addition, they are non-tox, non-allergen and have a docking score of - 10.83 kcal/mol, implying the best interaction with Pregnane X receptor involved in dysbiosis. The potential of nanosynbiotics to revolutionize treatment strategies through precise targeting and modulation of the gut microbiome for improved health outcomes and disease management is promising. Their transformational influence is projected to be powered by integration with modern technology and customized formulas. Further in-vivo studies are required for the validation of nanosynbiotics as nutraceuticals.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Artificial Intelligence , COVID-19 , SARS-CoV-2 , Drug Compounding/methods , Nanoparticles/chemistry , Inulin/chemistry
4.
Front Cell Infect Microbiol ; 14: 1430586, 2024.
Article in English | MEDLINE | ID: mdl-39170985

ABSTRACT

Background: Psoriasis is one of the most common autoimmune skin diseases. Increasing evidence shows that alterations in the diversity and function of microbiota can participate in the pathogenesis of psoriasis through various pathways and mechanisms. Objective: To review the connection between microbial changes and psoriasis, how microbial-targeted therapy can be used to treat psoriasis, as well as the potential of prebiotics, probiotics, synbiotics, fecal microbiota transplantation, diet, and Traditional Chinese Medicine as supplementary and adjunctive therapies. Methods: Literature related to the relationship between psoriasis and gut microbiota was searched in PubMed and CNKI. Results: Adjunct therapies such as dietary interventions, traditional Chinese medicine, and probiotics can enhance gut microbiota abundance and diversity in patients with psoriasis. These therapies stimulate immune mediators including IL-23, IL-17, IL-22, and modulate gamma interferon (IFN-γ) along with the NF-kB pathway, thereby suppressing the release of pro-inflammatory cytokines and ameliorating systemic inflammatory conditions. Conclusion: This article discusses the direction of future research and clinical treatment of psoriasis from the perspective of intestinal microbiota and the mechanism of traditional Chinese medicine, so as to provide clinicians with more comprehensive diagnosis and treatment options and bring greater hope to patients with psoriasis.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Medicine, Chinese Traditional , Probiotics , Psoriasis , Psoriasis/therapy , Psoriasis/microbiology , Psoriasis/drug therapy , Humans , Probiotics/therapeutic use , Prebiotics , Cytokines/metabolism , Interleukin-17/metabolism
5.
Carbohydr Polym ; 343: 122478, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174101

ABSTRACT

Lentinula edodes (Shiitake) is an important edible mushroom and polysaccharides are its major constituents with proven health benefits. The study was to investigate the gut bacterial fermentation and subsequent effects on gut barrier function of a glucan-rich polysaccharide, LePS40 precipitated from the mushroom water extract with 40 % (v/v) ethanol. LePS40 consisted of a ß-(1→3)-glucan main chain with substitution in the C-6 position with side chains mainly composed of (1 → 6)-linked ß-Glcp residues, (1 → 6)-linked α-Galp residues and terminal residues of ß-Glcp. LePS40 was found highly resistant to digestive enzymes and gastric acid in simulated human gastrointestinal tract, but highly fermentable during in vitro human fecal fermentation. The fecal fermentation degradation of LePS40 appeared to selectively break the glucoside linkage in view of the dramatic decrease in the glucose molar ratio (12.68 to 1.07). Compared with the prebiotic reference FOS, LePS40 led to much higher levels of butyric, and propionic acid and a lower level of acetic acid. Moreover, LePS40 enhanced the abundance of some beneficial bacterial populations, but decreased the bacteria possibly linked with fatty-liver disease and colorectal cancer. Furthermore, the fecal fermentation products of LePS40 showed a potential protective effect on intestinal barrier function against inflammatory damage in Caco-2/Raw264.7 co-culture model. These findings suggest the potential of LePS40 for improvement of gut health through modulation of gut microbiota.


Subject(s)
Fermentation , Gastrointestinal Microbiome , Shiitake Mushrooms , Shiitake Mushrooms/chemistry , Shiitake Mushrooms/metabolism , Humans , Gastrointestinal Microbiome/drug effects , Caco-2 Cells , Animals , Feces/microbiology , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/metabolism , Digestion/drug effects , Molecular Weight , Mice , Intestinal Mucosa/metabolism , Prebiotics
6.
Carbohydr Polym ; 343: 122442, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174123

ABSTRACT

Nanocellulose, a versatile and sustainable nanomaterial derived from cellulose fibers, has attracted considerable attention in various fields due to its unique properties. Similar to dietary fibers, nanocellulose is difficult to digest in the human gastrointestinal tract. The indigestible nanocellulose is fermented by gut microbiota, producing metabolites and potentially exhibiting prebiotic activity in intestinal diseases. Additionally, nanocellulose can serve as a matrix material for probiotic protection and show promising prospects for probiotic delivery. In this review, we summarize the classification of nanocellulose, including cellulose nanocrystals (CNC), cellulose nanofibers (CNF), and bacterial nanocellulose (BNC), highlighting their distinct characteristics and applications. We discuss the metabolism-related characteristics of nanocellulose from oral ingestion to colon fermentation and introduce the prebiotic activity of nanocellulose in intestinal diseases. Furthermore, we provide an overview of commonly used nanocellulose-based encapsulation techniques, such as emulsification, extrusion, freeze drying, and spray drying, as well as the delivery systems employing nanocellulose matrix materials, including microcapsules, emulsions, and hydrogels. Finally, we discuss the challenges associated with nanocellulose metabolism, prebiotic functionality, encapsulation techniques, and delivery systems using nanocellulose matrix material for probiotics. This review will provide new insight into the application of nanocellulose in the treatment of intestinal diseases and probiotic delivery.


Subject(s)
Cellulose , Gastrointestinal Microbiome , Nanoparticles , Probiotics , Cellulose/chemistry , Cellulose/metabolism , Humans , Nanoparticles/chemistry , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Animals , Nanofibers/chemistry , Fermentation , Prebiotics/administration & dosage , Drug Delivery Systems/methods
7.
BMC Gastroenterol ; 24(1): 283, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174901

ABSTRACT

BACKGROUND: There is a contradiction in the use of microbiota-therapies, including probiotics, prebiotics, and synbiotics, to improve the condition of patients with nonalcoholic fatty liver disease (NAFLD). The aim of this review was to evaluate the effect of microbiota-therapy on liver injury, inflammation, and lipid levels in individuals with NAFLD. METHODS: Using Pubmed, Embase, Cochrane Library, and Web of Science databases were searched for articles on the use of prebiotic, probiotic, or synbiotic for the treatment of patients with NAFLD up to March 2024. RESULTS: Thirty-four studies involving 12,682 individuals were included. Meta-analysis indicated that probiotic, prebiotic, and synbiotic supplementation significantly improved liver injury (hepatic fibrosis, SMD = -0.31; 95% CI: -0.53, -0.09; aspartate aminotransferase, SMD = -0.35; 95% CI: -0.55, -0.15; alanine aminotransferase, SMD = -0.48; 95% CI: -0.71, -0.25; alkaline phosphatase, SMD = -0.81; 95% CI: -1.55, -0.08), lipid profiles (triglycerides, SMD = -0.22; 95% CI: -0.43, -0.02), and inflammatory factors (high-density lipoprotein, SMD = -0.47; 95% CI: -0.88, -0.06; tumour necrosis factor alpha, SMD = -0.86 95% CI: -1.56, -0.56). CONCLUSION: Overall, supplementation with probiotic, prebiotic, or synbiotic had a positive effect on reducing liver enzymes, lipid profiles, and inflammatory cytokines in patients with NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Prebiotics , Probiotics , Synbiotics , Humans , Alanine Transaminase/blood , Inflammation , Lipids/blood , Liver , Non-alcoholic Fatty Liver Disease/therapy , Probiotics/therapeutic use , Randomized Controlled Trials as Topic , Synbiotics/administration & dosage
8.
PLoS One ; 19(8): e0307909, 2024.
Article in English | MEDLINE | ID: mdl-39159201

ABSTRACT

Foxtail millet (FM) and green banana (GB) are rich in health-promoting nutrients and bioactive substances, like antioxidants, dietary fibers, and various essential macro and micronutrients. Utilizing GB and FM flour as prebiotics is attributed to their ability to support gut health and offer multiple health benefits. The present study aimed to evaluate the impact of incorporating 10% GB flour (GBF) and different proportions (10-40%) of FM flour (FMF) on the prebiotic potential, antioxidant, nutrient, color, cooking quality, water activity and sensory attributes of noodles. The prebiotic potential, antioxidant, and nutrient of the produced noodles were significantly improved by increasing the levels of FMF. Sensorial evaluation revealed that noodles containing 30% FMF and 10% GBF attained comparable scores to the control sample. Furthermore, the formulated noodles exhibited significantly (p < 0.05) higher levels of protein, essential minerals (such as iron, magnesium, and manganese), dietary fiber (9.37 to 12.71 g/100 g), total phenolic compounds (17.81 to 36.35 mg GA eq./100 g), and total antioxidants (172.57 to 274.94 mg AA eq./100 g) compared to the control. The enriched noodles also demonstrated substantially (p < 0.05) increased antioxidant capacity, as evidenced by enhanced DPPH and FRAP activities, when compared to the control noodles. Overall, the incorporation of 30% FMF and 10% GBF led to a noteworthy improvement in the nutritional and antioxidant qualities of the noodles, as well as the prebiotic potential of the noodles with regard to L. plantarum, L. rhamnosus, and L. acidophilus. The implementation of this enrichment strategy has the potential to confer a multitude of health advantages.


Subject(s)
Antioxidants , Flour , Musa , Nutritive Value , Prebiotics , Antioxidants/analysis , Prebiotics/analysis , Flour/analysis , Musa/chemistry , Dietary Fiber/analysis , Millets/chemistry , Humans
9.
Compr Rev Food Sci Food Saf ; 23(5): e13431, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39165140

ABSTRACT

Human milk oligosaccharides (HMOs) are an evolutionarily significant advantage bestowed by mothers for facilitating the development of the infant's gut microbiota. They can avoid absorption in the stomach and small intestine, reaching the colon successfully, where they engage in close interactions with gut microbes. This process also enables HMOs to exert additional prebiotic effects, including regulating the mucus layer, promoting physical growth and brain development, as well as preventing and mitigating conditions such as NEC, allergies, and diarrhea. Here, we comprehensively review the primary ways by which gut microbiota, including Bifidobacteria and other genera, utilize HMOs, and we classify them into five central pathways. Furthermore, we emphasize the metabolic benefits of bacteria consuming HMOs, particularly the recently identified intrinsic link between HMOs and the metabolic conversion of tryptophan to indole and its derivatives. We also examine the extensive probiotic roles of HMOs and their recent research advancements, specifically concentrating on the unsummarized role of HMOs in regulating the mucus layer, where their interaction with the gut microbiota becomes crucial. Additionally, we delve into the principal tools used for functional mining of new HMOs. In conclusion, our study presents a thorough analysis of the interaction mechanism between HMOs and gut microbiota, emphasizing the cooperative utilization of HMOs by gut microbiota, and provides an overview of the subsequent probiotic effects of this interaction. This review provides new insights into the interaction of HMOs with the gut microbiota, which will inform the mechanisms by which HMOs function.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Oligosaccharides , Prebiotics , Humans , Gastrointestinal Microbiome/physiology , Milk, Human/chemistry , Milk, Human/microbiology , Oligosaccharides/chemistry , Probiotics , Infant , Bacteria/metabolism , Bifidobacterium/physiology
10.
J Agric Food Chem ; 72(33): 18497-18506, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39099138

ABSTRACT

Synbiotics, the combination of probiotics and prebiotics, are thought to be a pragmatic approach for the treatment of various diseases, including inflammatory bowel disease (IBD). The synergistic therapeutic effects of probiotics and prebiotics remain underexplored. Clostridium tyrobutyricum, a short-chain fatty acid (SCFA) producer, has been recognized as a promising probiotic candidate that can offer health benefits. In this study, the treatment effects of synbiotics containing C. tyrobutyricum and chitooligosaccharides (COSs) on IBD were evaluated. The results indicated that the synbiotic supplement effectively relieved inflammation and restored intestinal barrier function. Additionally, the synbiotic supplement could contribute to the elimination of reactive oxygen species (ROS) and improve the production of SCFAs through the SCFAs-producer of C. tyrobutyricum. Furthermore, such the synbiotic could also regulate the composition of gut microbiota. These findings underscore the potential of C. tyrobutyricum and COSs as valuable living biotherapeutics for the treatment of intestinal-related diseases.


Subject(s)
Clostridium tyrobutyricum , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Oligosaccharides , Synbiotics , Gastrointestinal Microbiome/drug effects , Oligosaccharides/administration & dosage , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/metabolism , Clostridium tyrobutyricum/metabolism , Animals , Humans , Synbiotics/administration & dosage , Mice , Male , Fatty Acids, Volatile/metabolism , Mice, Inbred C57BL , Probiotics/administration & dosage , Probiotics/pharmacology , Prebiotics/administration & dosage , Chitosan
11.
Microbiol Res ; 287: 127857, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39121703

ABSTRACT

The microbiota in the female genital tract is an intricate assembly of diverse aerobic, anaerobic, and microaerophilic microorganisms, which share the space within the reproductive tract and engage in complex interactions. Microbiome dysbiosis may disrupt the symbiotic relationship between the host and microorganisms and play a pivotal role in the pathogenesis of various diseases, including its involvement in the establishment of human papillomavirus (HPV)-associated cervical cancer (CC). Interventions to restore microbiota homeostasis (e.g., probiotics) and bacterial-vector HPV therapeutic vaccines have been reported to be potentially effective in clearing HPV infection and ameliorating cytological abnormalities. In this review, we place emphasis on elucidating the alterations within the cervical-vaginal microbiota as well as the intratumoral microbiota in the context of high-risk HPV (HR-HPV) infection and its subsequent progression to cervical intraepithelial neoplasia/CC. Furthermore, we explore the mechanisms by which these microbial communities exert potential pathogenic or protective effects, including modulating genital inflammation and immune responses, affecting HR-HPV oncogene expression and oncoprotein production, regulating oxidative stress and deoxyribonucleic acid (DNA) damage, and inducing metabolic rewiring. Lastly, we summarize the latest evidence in human trials regarding the efficacy of probiotics, prebiotics and probiotic-vector HPV therapeutic vaccines. This review aims to foster a deeper understanding of the role of the microbiota in HR-HPV infection-related cervix cancer development, and further provide a theoretical basis for the development of preventive and therapeutic strategies based on microbial modulation.


Subject(s)
Dysbiosis , Microbiota , Papillomavirus Infections , Probiotics , Uterine Cervical Neoplasms , Vagina , Humans , Female , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/microbiology , Uterine Cervical Neoplasms/therapy , Papillomavirus Infections/virology , Papillomavirus Infections/therapy , Vagina/microbiology , Vagina/virology , Probiotics/therapeutic use , Probiotics/administration & dosage , Papillomaviridae/pathogenicity , Papillomaviridae/physiology , Cervix Uteri/microbiology , Cervix Uteri/virology , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Dysplasia/virology , Uterine Cervical Dysplasia/microbiology , Uterine Cervical Dysplasia/therapy , Prebiotics/administration & dosage
12.
Sci Rep ; 14(1): 18866, 2024 08 14.
Article in English | MEDLINE | ID: mdl-39143192

ABSTRACT

Isomalto/malto-polysaccharides (IMMPs) are a novel type of soluble dietary fibres with a prebiotic potential promoting growth of beneficial microbes in the gut. However, the mode of action of IMMPs remains unknown. Previous studies on IMMPs showed an increase in total bacteria, especially lactobacilli, and higher production of short chain fatty acids (SCFA) when IMMPs were fed to rats or used during in vitro fermentation. Here we used metatranscriptomics to investigate how IMMPs with different amounts of α - (1 → 6) glycosidic linkages affected microbial function during incubation with human fecal inoculum. We showed that active microbial community dynamics during fermentation varied depending on the type of IMMP used and that the observed changes were reflected in the community gene expression profiles. Based on metatranscriptome analysis, members of Bacteroides, Lactobacillus and Bifidobacterium were the predominant degraders of IMMPs, and the increased gene expression in these bacteria correlated with high amounts of α - (1 → 6) glycosidic linkages. We also noted an increase in relative abundance of these bacteria and an activation of pathways involved in SCFA synthesis. Our findings could provide a baseline for more targeted approaches in designing prebiotics for specific bacteria and to achieve more controlled modulation of microbial activity towards desired health outcomes.


Subject(s)
Feces , Gastrointestinal Microbiome , Prebiotics , Humans , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Fermentation , Polysaccharides/metabolism , Colon/microbiology , Colon/metabolism , Fatty Acids, Volatile/metabolism , Transcriptome , Gene Expression Profiling , Dietary Fiber/metabolism , Lactobacillus/metabolism , Lactobacillus/genetics , Bacteria/genetics , Bacteria/metabolism
13.
Med Oncol ; 41(9): 231, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39162936

ABSTRACT

Colorectal cancer (CRC), the third most common cancer worldwide, develops mainly due to the accumulation of genetic and epigenetic changes over many years. Substantial evidence suggests that gut microbiota plays a significant role in the initiation, progression, and control of CRC, depending on the balance between beneficial and pathogenic microorganisms. Nonetheless, gut microbiota composition by regulating the host immune response may either promote or inhibit CRC. Thus, modification of gut microbiota potentially impacts clinical outcomes of immunotherapy. Previous studies have indicated that therapeutic strategies such as probiotics, prebiotics, and postbiotics enhance the intestinal immune system and improve the efficacy of immunotherapeutic agents, potentially serving as a complementary strategy in cancer immunotherapy. This review discusses the role of the gut microbiota in the onset and development of CRC in relation to the immune response. Additionally, we focus on the effect of strategies manipulating gut microbiome on the immune response and efficacy of immunotherapy against CRC. We demonstrate that manipulation of gut microbiome can enhance immune response and outcomes of immunotherapy through downregulating Treg cells and other immunosuppressive cells while improving the function of T cells within the tumor; however, further research, especially clinical trials, are needed to evaluate its efficacy in cancer treatment.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Immunomodulation , Immunotherapy , Probiotics , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/therapy , Gastrointestinal Microbiome/immunology , Probiotics/therapeutic use , Immunomodulation/immunology , Immunotherapy/methods , Prebiotics/administration & dosage
14.
Int J Biol Macromol ; 277(Pt 1): 133991, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089904

ABSTRACT

Galactooligosaccharides (GOS), as mimics of human milk oligosaccharides, are important prebiotics for modulating the ecological balance of intestinal microbiota. A novel carrier-free cell immobilization method was established using genipin to cross-link Kluyveromyces lactis CGMCC 2.1494, which produced ß-galactosidase, an enzyme essential for GOS synthesis. The resulting immobilized cells were characterized as stable by thermogravimetric analysis and confirmed to be crosslinked through scanning electron microscopy analysis (SEM) and Fourier transform infrared spectroscopy (FTIR). The Km and Vmax values of ß-galactosidase in immobilized cells towards o-nitrophenyl ß-D-galactoside were determined to be 3.446 mM and 2210 µmol min-1 g-1, respectively. The enzyme in the immobilized showed higher thermal and organic solvent tolerance compared to that in free cells. The immobilized cells were subsequently employed for GOS synthesis using plant-derived galactose as the substrate. The synthetic reaction conditions were optimized through both single-factor experiments and response surface methodology, resulting in a high yield of 49.1 %. Moreover, the immobilized cells showed good reusability and could be reused for at least 20 batches of GOS synthesis, with the enzyme activity remaining above 70 % at 35 °C.


Subject(s)
Cells, Immobilized , Galactose , Iridoids , Kluyveromyces , Oligosaccharides , Prebiotics , beta-Galactosidase , Iridoids/chemistry , Iridoids/metabolism , Galactose/chemistry , Oligosaccharides/chemistry , Cells, Immobilized/metabolism , Kluyveromyces/metabolism , beta-Galactosidase/metabolism , Cross-Linking Reagents/chemistry
15.
Pharmacol Res ; 207: 107334, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103131

ABSTRACT

The intestinal microbiota represents a key element in maintaining the homeostasis and health conditions of the host. Vascular pathologies and other risk factors such as aging have been recently associated with dysbiosis. The qualitative and quantitative alteration of the intestinal microbiota hinders correct metabolic homeostasis, causing structural and functional changes of the intestinal wall itself. Impairment of the intestinal microbiota, combined with the reduction of the barrier function, worsen the pathological scenarios of peripheral tissues over time, including the vascular one. Several experimental evidence, collected in this review, describes in detail the changes of the intestinal microbiota in dysbiosis associated with vascular alterations, such as atherosclerosis, hypertension, and endothelial dysfunction, the resulting metabolic disorders and how these can impact on vascular health. In this context, the gut-vascular axis is considered, for the first time, as a merged unit involved in the development and progression of vascular pathologies and as a promising target. Current approaches for the management of dysbiosis such as probiotics, prebiotics and dietary modifications act mainly on the intestinal district. Postbiotics, described as preparation of inanimate microorganisms and/or their components that confers health benefits on the host, represent an innovative strategy for a dual management of intestinal dysbiosis and vascular pathologies. In this context, this review has the further purpose of defining the positive effects of the supplementation of bacterial strains metabolites (short­chain fatty acids, exopolysaccharides, lipoteichoic acids, gallic acid, and protocatechuic acid) restoring intestinal homeostasis and acting directly on the vascular district through the gut-vascular axis.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Vascular Diseases , Humans , Gastrointestinal Microbiome/drug effects , Animals , Dysbiosis/microbiology , Vascular Diseases/microbiology , Probiotics/therapeutic use , Prebiotics/administration & dosage
16.
Phytomedicine ; 133: 155919, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153277

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a major clinical and global public health issue, with no specific pharmacological treatment available. Currently, there is a lack of approved drugs for the clinical treatment of NAFLD. Large-leaf yellow tea polysaccharides (YTP) is a natural biomacromolecule with excellent prebiotic properties and significant therapeutic effects on multiple metabolic diseases. However, the specific mechanisms by which YTP regulates NAFLD remain unclear. PURPOSE: This study aims to explore the prebiotic effects of YTP and the potential mechanisms by which it inhibits hepatic cholesterol accumulation in NAFLD mice. METHODS: The effects of YTP on lipid accumulation were evaluated in NAFLD mice through obesity trait analysis and bile acids (BAs) metabolism assessment. Additionally, fecal microbiota transplantation (FMT) was performed, and high-throughput sequencing was employed to investigate the mechanisms underlying YTP's regulatory effects on gut microbiota and BA metabolism. RESULTS: Our study demonstrated that YTP altered the constitution of colonic BA, particularly increasing the levels of conjugated BA and non-12OH BA, which suppressed ileum FXR receptors and hepatic BA reabsorption, facilitated BA synthesis, and fecal BA excretion. The modifications were characterized by a decrease in the levels of FXR, FGF15, FGFR4, and ASBT proteins, and an increase in the levels of Cyp7a1 and Cyp27a1 proteins. YTP might affect enterohepatic circulation and by the activated the hepatic FXR-SHP pathway. Meanwhile, YTP reshaped the intestinal microbiome structure by decreasing BSH-producing genera and increasing taurine metabolism genera. The correlation analysis implied that Muribaculaceae, Pseudomonas, acterium_coprostanoligenes_group, Clostridiales, Lachnospiraceae_NK4A136_group, Delftia, Dubosiella, and Romboutsia were strongly correlated with specific BA monomers. CONCLUSIONS: YTP modulates bile salt hydrolase-related microbial genera to activate alternative bile acid synthesis pathways, thereby inhibiting NAFLD progression. These results suggest that YTP may serve as a potential probiotic formulation, offering a feasible dietary intervention for NAFLD.


Subject(s)
Bile Acids and Salts , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Polysaccharides , Tea , Animals , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/metabolism , Gastrointestinal Microbiome/drug effects , Bile Acids and Salts/metabolism , Polysaccharides/pharmacology , Male , Mice , Tea/chemistry , Liver/drug effects , Liver/metabolism , Prebiotics , Fibroblast Growth Factors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Amidohydrolases/metabolism
17.
Microb Pathog ; 194: 106844, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39128644

ABSTRACT

This study investigated the effect of pumpkin powder (2 %, 4 %, and 6 %) and Enterococcus faecium and Enterococcus faecalis probiotics on the physicochemical, microbiological, and sensory properties of yogurt samples during 28 days of storage at 4 °C. The prebiotic effect of pumpkin powder (Cucurbita pepo) and the probiotic effect of Enterococcus faecium and E. faecalis were determined. Adding pumpkin powder to yogurt did not significantly alter the pH, acidity, fat, protein, and ash content (p > 0.05). Water holding was not changed during the storage time in the samples of probiotic yogurts, but as the pumpkin powder content increased, the water holding capacity also increased (p < 0.05). This situation did lead to a reduction in syneresis (p < 0.05). The lowest gumminess value at the end of storage was found in the D2 sample (p < 0.05), and the highest adhesiveness value was found in the D4 sample (p < 0.05). Furthermore, throughout the 28-day storage period, E. faecium and E. faecalis maintained a live cell count of ≥6 log CFU g-1 in the probiotic product. As a result of the statistical evaluation, there was a decrease in E. faecium in the D4, S2, and S4 samples, and then it increased again (p > 0.05) during the storage time. As a result of the statistical evaluation, it was determined that the smell, consistency in the spoon, consistency in the mouth, flavor, and acidity changes during the storage were not substantial (p > 0.05). In conclusion, it was found that pumpkin, a byproduct of the pumpkin seed industry, has the potential to act as a prebiotic and improve the properties of dairy products. Additionally, the study suggests that E. faecium and E. faecalis strains could be suitable for probiotic yogurts.


Subject(s)
Cucurbita , Enterococcus faecalis , Enterococcus faecium , Prebiotics , Probiotics , Yogurt , Enterococcus faecium/growth & development , Cucurbita/microbiology , Enterococcus faecalis/growth & development , Enterococcus faecalis/drug effects , Yogurt/microbiology , Hydrogen-Ion Concentration , Food Microbiology , Food Storage , Colony Count, Microbial , Taste
18.
Altern Ther Health Med ; 30(7): 6-10, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39110053

ABSTRACT

Contenxt: The focus on gastrointestinal (GI) health has been increasing due to a growing awareness of its pivotal role in overall well-being. Current trends in addressing GI health aim to optimize gut-microbiome composition, understand the bidirectional communication between the gut and the brain, and use functional diagnostic testing to aid in diagnosis and guide treatments for GI disorders. Objective: The study intended to examine advances in microbiome research, including advancements in technology, and to investigate methods of noninvasive diagnostics, the importance of the gut-brain axis (GBA) and the oral cavity, the types of beneficial microorganisms, and the prevalence of functional GI disorders (FGIDs). Setting: The study took place at Biocidin Botanicals in Watsonville CA, USA. Results: Recent research has illuminated the critical role that gut health plays in human physiology and disease. Also, the oral cavity is emerging as a vital microbial reservoir affecting both GI and systemic health. Dietary patterns and lifestyle choices exert profound effects on the gut microbiota and host metabolism, emphasizing the importance of holistic approaches to GI care. FGIDs impose significant burdens on patients' quality of life and the healthcare systems. Integrating stress management strategies and supporting healthy lifestyle choices are essential for managing FGIDs effectively. Conclusions: Functional foods, prebiotics, probiotics, and postbiotics offer promising avenues for optimizing GI health and mitigating disease risk.


Subject(s)
Gastrointestinal Diseases , Gastrointestinal Microbiome , Humans , Gastrointestinal Diseases/therapy , Probiotics/therapeutic use , Brain-Gut Axis/physiology , Prebiotics
19.
Acc Chem Res ; 57(16): 2234-2244, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39115809

ABSTRACT

ConspectusThe origin of the single chirality of the chemical building blocks of life remains an intriguing topic of research, even after decades of experimental and theoretical work proposing processes that may break symmetry and induce chiral amplification, a term that may be defined as the enhancement of enantiomeric excess starting from prochiral substrates or from a racemic mixture or a small imbalance between enantiomers. Studies aimed at understanding prebiotically plausible pathways to these molecules have often neglected the issue of chirality, with a focus on the stereochemical direction of these reactions generally being pursued after reaction discovery. Our work has explored how the stereochemical outcome for the synthesis of amino acids and sugars might be guided to rationalize the origin of biological homochirality. The mechanistic interconnection between enantioenrichment in these two groups of molecules provides insights concerning the handedness extant in modern biology. In five separate examples involving the synthesis of life's building blocks, including sugars, RNA precursors, amino acids, and peptides, kinetic resolution emerges as a key protocol for enantioenrichment from racemic molecules directed by chiral source molecules. Several of these examples involve means not only for chiral amplification but also symmetry breaking and chirality transfer across a range of racemic monomer molecules. Several important implications emerge from these studies: one, kinetic resolution of the primordial chiral sugar, glyceraldehyde, plays a key role in a number of different prebiotically plausible reactions; two, the emergence of homochirality in sugars and amino acids is inherently intertwined, with clear synergy between the biological hand of each molecule class; three, the origin story for the homochirality of enzymes and modern metabolism points toward kinetic resolution of racemic amino acids in networks that later evolved to include sophisticated and complete catalytic and co-catalytic cycles; four, a preference for heterochiral ligation forming product molecules that cannot lead to biologically competent polymers can in fact be a driving force for a route to homochiral polymer chains; and five, enantioenrichment in complex mixtures need not be addressed one compound at a time, because kinetic resolution induces symmetry breaking and chirality transfer that may lead to general protocols rather than specific cases tailored to each individual molecule. Such chirality transfer mechanisms perhaps presage strategies utilized in modern biology.Our latest work extends the study of monomer enantioenrichment to the ligation of these molecules into the extended homochiral chains leading to the complex polymers of modern biology. A central theme in all of these reactions is the key role that kinetic resolution of a racemic mixture of amino acids or sugars plays in enabling enantioenrichment under prebiotically plausible conditions. This work has uncovered important trends in symmetry breaking, chirality transfer, and chiral amplification. Kinetic resolution of racemic mixtures emerges as a general solution for chiral amplification in prebiotic chemistry, leading to the single chirality of complex biological molecules and genetic polymers.


Subject(s)
Amino Acids , Stereoisomerism , Kinetics , Amino Acids/chemistry , Peptides/chemistry , Peptides/chemical synthesis , Prebiotics , Origin of Life , Sugars/chemistry , RNA/chemistry
20.
Nutrients ; 16(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125448

ABSTRACT

It is now established that patients with rheumatoid arthritis (RA) have an increased risk of developing cervical cancer (CC) or its precursor, cervical intraepithelial neoplasia (CIN). However, the underlying mechanisms of this association have not been elucidated. RA is characterized by unresolved chronic inflammation. It is suggested that human papillomavirus (HPV) infection in RA patients exacerbates inflammation, increasing the risk of CC. The tumor microenvironment in RA patients with CC is also marked by chronic inflammation, which aggravates the manifestations of both conditions. Gut and vaginal dysbiosis are also considered potential mechanisms that contribute to the chronic inflammation and aggravation of RA and CC manifestations. Numerous clinical and pre-clinical studies have demonstrated the beneficial effects of various nutritional approaches to attenuate chronic inflammation, including polyunsaturated fatty acids and their derivatives, specialized pro-resolving mediators (SPMs), probiotics, prebiotics, and certain diets. We believe that successful resolution of chronic inflammation and correction of dysbiosis, in combination with current anti-RA and anti-CC therapies, is a promising therapeutic approach for RA and CC. This approach could also reduce the risk of CC development in HPV-infected RA patients.


Subject(s)
Arthritis, Rheumatoid , Dysbiosis , Papillomavirus Infections , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/therapy , Arthritis, Rheumatoid/complications , Female , Dysbiosis/complications , Papillomavirus Infections/complications , Probiotics/therapeutic use , Inflammation , Gastrointestinal Microbiome , Prebiotics , Tumor Microenvironment , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL