Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27.189
Filter
1.
Front Cell Infect Microbiol ; 14: 1354736, 2024.
Article in English | MEDLINE | ID: mdl-39045133

ABSTRACT

The present study evaluated the capacity of three Bacillus species to improve health status and growth performance of Nile Tilapia fed with high levels of soybean meal and challenged with Aeromonas hydrophila. In vitro experiments showed that ß-hemolysin and metalloprotease enzymes were produced by A. hydrophila throughout the exponential growth phase. In vivo experiments showed that 107 colony-forming units (CFUs)/ml of this pathogen killed 50% of control group fishes in 13 days. To evaluate the influence of Bacillus strains on health status and growth performance in Nile Tilapia, 180 fishes (33.44 + 0.05 g) were distributed in 12 tanks of 200 L each, and animals were fed twice per day until satiety. 1) Control group without Bacillus, 2) Bacillus sp1, 3) Bacillus sp2, and 4) Bacillus sp3 groups were formulated containing 106 CFU/g. After 40 days of feeding, the fishes were intraperitoneally injected with 1 ml of A. hydrophila at 2 × 107 CFU/ml, and mortality was recorded. The results showed that cumulative mortality rate was significantly (p< 0.05) lower in the Bacillus sp1 (25%), sp2 (5%), and sp3 (15%) groups, than the control group (50%). Weight gain was also significantly better (p< 0.05) in the Bacillus sp1 (36%), sp2 (67%), and sp3 (55%) groups with respect to the control group (30%). In conclusion, functional diet formulated with high levels of soybean meal and supplemented with Bacillus sp2 could be an alternative to protect Nile tilapia cultures from A. hydrophila infections and improve fish growth performance.


Subject(s)
Aeromonas hydrophila , Bacillus , Cichlids , Fish Diseases , Gram-Negative Bacterial Infections , Animals , Fish Diseases/microbiology , Fish Diseases/prevention & control , Cichlids/growth & development , Cichlids/microbiology , Aeromonas hydrophila/pathogenicity , Aeromonas hydrophila/growth & development , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/veterinary , Gram-Negative Bacterial Infections/prevention & control , Animal Feed , Probiotics/administration & dosage , Glycine max/microbiology , Aquaculture
2.
Gut Microbes ; 16(1): 2379566, 2024.
Article in English | MEDLINE | ID: mdl-39013030

ABSTRACT

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants with no specific treatments available. We aimed to identify the molecular mechanisms underlying NEC and investigate the therapeutic effects of Bacteroides fragilis on NEC. Clinical samples of infant feces, bile acid-targeted metabolomics, pathological staining, bioinformatics analysis, NEC rat model, and co-immunoprecipitation were used to explore the pathogenesis of NEC. Taxonomic characterization of the bile salt hydrolase (bsh) gene, enzyme activity assays, 16S rRNA sequencing, and organoids were used to explore the therapeutic effects of B. fragilis on NEC-related intestinal damage. Clinical samples, NEC rat models, and in vitro experiments revealed that total bile acid increased in the blood but decreased in feces. Moreover, the levels of FXR and other bile acid metabolism-related genes were abnormal, resulting in disordered bile acid metabolism in NEC. Taurochenodeoxycholic acid accelerated NEC pathogenesis and taurodeoxycholate alleviated NEC. B. fragilis displayed bsh genes and enzyme activity and alleviated intestinal damage by restoring gut microbiota dysbiosis and bile acid metabolism abnormalities by inhibiting the FXR-NLRP3 signaling pathway. Our results provide valuable insights into the therapeutic role of B. fragilis in NEC. Administering B. fragilis may substantially alleviate intestinal damage in NEC.


Subject(s)
Amidohydrolases , Bacteroides fragilis , Bile Acids and Salts , Enterocolitis, Necrotizing , Gastrointestinal Microbiome , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/drug therapy , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Bacteroides fragilis/metabolism , Bacteroides fragilis/genetics , Signal Transduction/drug effects , Bile Acids and Salts/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Gastrointestinal Microbiome/drug effects , Amidohydrolases/metabolism , Amidohydrolases/genetics , Humans , Rats, Sprague-Dawley , Infant, Newborn , Disease Models, Animal , Male , Female , Probiotics/administration & dosage , Probiotics/pharmacology , Infant, Premature , Dysbiosis/microbiology
3.
FASEB J ; 38(14): e23801, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39018106

ABSTRACT

Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.


Subject(s)
Keratinocytes , Lactobacillus , Staphylococcus aureus , Humans , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/drug effects , Skin/microbiology , Skin/metabolism , Wound Healing/drug effects , Probiotics/pharmacology , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Ribonucleases/metabolism
4.
Nanoscale ; 16(28): 13399-13406, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38953700

ABSTRACT

Living drugs offer a new frontier in medicine, paving the way for personalized and potentially curative treatments. A customized living drug generally requires specialized technologies for highly effective and selective delivery to lesion locations. In this study, we explored an interfacial engineering method for living drugs by wrapping them with a "stealth coating", achieving "ON/OFF" switching of the communications between probiotics and the gastrointesinal (GI) tract. This maximized the bioactivity of living drugs following oral administration to exempt acidic insults and then significantly improved the retention through the gastrointestinal tract. With the notable ability to improve oral availability, the interfacial-engineered living drugs represent remarkable effects for enhanced oral delivery and treatment efficacy in the dextran sulfate sodium (DSS)-induced acute colitis model. We believe that this work has the potential to revolutionize medicine by precisely targeting and increasing curative activity in the future of disease treatment.


Subject(s)
Colitis , Dextran Sulfate , Probiotics , Administration, Oral , Animals , Probiotics/chemistry , Probiotics/administration & dosage , Mice , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/chemistry , Humans , Drug Delivery Systems
5.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38977897

ABSTRACT

Sleeplessness (insomnia) is a potential symptom of depression. A probiotic NVP1704 alleviates depression-like behavior and neuroinflammation in mice. Therefore, to understand whether NVP1704 could be effective against sleeplessness in vivo, we exposed immobilization stress (IS) in mice, then orally administered NVP1704 for 5 days, and assayed depression/anxiety-like behavior in the open field, elevated plus maze, and tail suspension tests, sleeping latency time, and sleep duration, euthanized then by exposure to CO2, and analyzed their related biomarkers. Oral administration of NVP1704 decreased IS-induced depression/anxiety-like behavior and sleeping latency time and increased IS-suppressed sleeping duration. NVP1704 increased IS-suppressed expression of γ-aminobutyric acid (GABA), GABAA receptor α1 (GABAARα1) and α2 subunits (GABAARα2), serotonin, 5-HT receptors (5-HT1AR and 5-HT1BR), and melatonin receptors (MT1R and MT2R) in the prefrontal cortex and thalamus. NVP1704 also increased the IS-suppressed GABAARα1-positive cell population in the prefrontal cortex and decreased IS-induced corticosterone, TNF-α, and IL-6 expression and the NF-κB+Iba1+ cell population in the brain and myeloperoxidase, TNF-α, and IL-6 expression and the NF-κB+CD11c+ cell population in the colon. Based on these findings, NVP1704 may alleviate depression/anxiety/sleeplessness-like behaviors through the upregulation of serotonergic and GABAergic systems and downregulation of NF-κB activation.


Subject(s)
Depression , NF-kappa B , Probiotics , Animals , Mice , Probiotics/administration & dosage , Probiotics/pharmacology , NF-kappa B/metabolism , Depression/etiology , Depression/drug therapy , Depression/metabolism , Male , Serotonin/metabolism , gamma-Aminobutyric Acid/metabolism , Stress, Psychological/drug therapy , Down-Regulation , Up-Regulation , Receptors, Serotonin/metabolism , Receptors, Serotonin/genetics
6.
Mar Drugs ; 22(7)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39057417

ABSTRACT

To improve probiotics' survivability during gastrointestinal digestion and heat treatment, Lactobacillus plantarum was microencapsulated by spray-drying using Laminaria japonica polysaccharide/sodium caseinate/gelatin (LJP/SC/GE) composites. Thermogravimetry and differential scanning calorimetry results revealed that the denaturation of LJP/SC/GE microcapsules requires higher thermal energy than that of SC/GE microcapsules, and the addition of LJP may improve thermal stability. Zeta potential measurements indicated that, at low pH of the gastric fluid, the negatively charged LJP attracted the positively charged SC/GE, helping to maintain an intact microstructure without disintegration. The encapsulation efficiency of L. plantarum-loaded LJP/SC/GE microcapsules reached about 93.4%, and the survival rate was 46.9% in simulated gastric fluid (SGF) for 2 h and 96.0% in simulated intestinal fluid (SIF) for 2 h. In vitro release experiments showed that the LJP/SC/GE microcapsules could protect the viability of L. plantarum in SGF and release probiotics slowly in SIF. The cell survival of LJP/SC/GE microcapsules was significantly improved during the heat treatment compared to SC/GE microcapsules and free cells. LJP/SC/GE microcapsules can increase the survival of L. plantarum by maintaining the lactate dehydrogenase and Na+-K+-ATPase activity. Overall, this study demonstrates the great potential of LJP/SC/GE microcapsules to protect and deliver probiotics in food and pharmaceutical systems.


Subject(s)
Capsules , Hot Temperature , Lactobacillus plantarum , Laminaria , Polysaccharides , Laminaria/chemistry , Polysaccharides/pharmacology , Polysaccharides/chemistry , Probiotics/pharmacology , Probiotics/administration & dosage , Digestion/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Hydrogen-Ion Concentration , Gelatin/chemistry , Gelatin/pharmacology , Microbial Viability/drug effects , Edible Seaweeds
7.
BMC Complement Med Ther ; 24(1): 258, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987744

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal system. So far, no treatment has been identified that can completely cure IBD. Lactobacillus brevis is hypothesized to be beneficial in preventing inflammation. This study aimed to evaluate the potential probiotic effects of live and pasteurized L. brevis IBRC-M10790 on the in vitro cell co-culture model of IBD. METHODS: An in vitro intestinal model was established using a transwell co-culture system of Caco-2 intestinal epithelial cells and RAW264.7 macrophages. Inflammatory conditions were induced in RAW264.7 cells using lipopolysaccharide. The effects of live and pasteurized L. brevis IBRC-M10790 on inflammatory mediators and epithelial barrier markers were investigated. RESULTS: L. brevis IBRC-M10790 was able to significantly decrease the proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) and increase the anti-inflammatory cytokine (IL-10) in the in vitro co-culture system. In addition, L. brevis increased adherens and tight junction (TJ) markers (ZO-1, E-cadherin, and Occludin) in Caco-2 intestinal epithelial cells. Based on the results, pasteurized L. brevis showed a higher protective effect than live L. brevis. CONCLUSIONS: Our findings suggest that live and pasteurized forms of L. brevis possess probiotic properties and can mitigate inflammatory conditions in IBD.


Subject(s)
Anti-Inflammatory Agents , Inflammatory Bowel Diseases , Levilactobacillus brevis , Probiotics , Probiotics/pharmacology , Humans , Caco-2 Cells , Inflammatory Bowel Diseases/drug therapy , Mice , Animals , RAW 264.7 Cells , Anti-Inflammatory Agents/pharmacology , Coculture Techniques , Cytokines/metabolism , Pasteurization
8.
Helicobacter ; 29(4): e13108, 2024.
Article in English | MEDLINE | ID: mdl-39021274

ABSTRACT

BACKGROUND: Helicobacter pylori infection-associated gastric adenocarcinoma is influenced by various factors, including the digestive microbiota. Lactic acid bacteria role in digestive carcinogenesis has been discussed, and some Lactobacillaceae family species have been shown to act against H. pylori-induced inflammation and colonization. However, their effects on H. pylori-related carcinogenesis have not yet been studied. Lactobacillaceae family effects on the epithelial-to-mesenchymal transition (EMT), emergence of cells with cancer stem cell (CSC) properties and the pro-inflammatory response of gastric epithelial cells to H. pylori infection were investigated. MATERIALS AND METHODS: A co-culture model of AGS gastric epithelial cells infected with a carcinogenic strain of H. pylori associated with 18 different probiotic strains candidates were used. Different EMT indicators and CSC properties were studied, including quantification of the mesenchymal phenotype, tumorsphere formation, EMT marker expression, and tight junction evaluation with immunofluorescence microscopy. The effect of the strains on the pro-inflammatory response to H. pylori was also evaluated by quantifying interleukin-8 (IL-8) production using ELISA. RESULTS: Among the strains tested, Lactobacillus gasseri BIO6369 and Lacticaseibacillus rhamnosus BIO5326 induced a 30.6% and 38.4% reduction in the mesenchymal phenotype, respectively, caused a significant decrease in Snail and Zeb1 EMT marker expression and prevented the loss of tight junctions induced by H. pylori infection. A separate co-culture with a Boyden chamber maintained the effects induced by the two strains. H. pylori-induced IL-8 production was also significantly reduced in the presence of L. gasseri BIO6369 and L. rhamnosus BIO5326. CONCLUSION: Lactobacillus gasseri BIO6369 and L. rhamnosus BIO5326 strains decreased epithelial-to-mesenchymal transition and inflammation induced by H. pylori infection, suggesting that these species may have a protective effect against H. pylori-induced gastric carcinogenesis.


Subject(s)
Epithelial Cells , Epithelial-Mesenchymal Transition , Helicobacter Infections , Helicobacter pylori , Lacticaseibacillus rhamnosus , Lactobacillus gasseri , Probiotics , Stomach Neoplasms , Humans , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/physiology , Helicobacter pylori/pathogenicity , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Lacticaseibacillus rhamnosus/physiology , Epithelial Cells/microbiology , Coculture Techniques , Carcinogenesis
9.
Benef Microbes ; 15(4): 411-415, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38960392

ABSTRACT

Clinical guidelines are recommendations for healthcare providers regarding patients with specific conditions. These guidelines should be based on practice experience and the best available research evidence. However, guidelines developed by various health organisations worldwide often do not agree with each other. This is also true for the current guidelines for the clinical use of probiotics. This article aims to provide examples of conflicting clinical guidelines for probiotics, define reasons for this phenomenon, describe standard tools for improving their quality, and suggest ways to enhance the development and assessment of suitable clinical guidelines for the appropriate clinical use of probiotics in specific conditions.


Subject(s)
Practice Guidelines as Topic , Probiotics , Probiotics/administration & dosage , Probiotics/therapeutic use , Probiotics/standards , Humans
10.
Nutrients ; 16(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38999758

ABSTRACT

Globally, metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty liver disease (NAFLD), is one of the most common liver disorders and is strongly associated with copper deficiency. To explore the potential effects and mechanisms of Lactiplantibacillus plantarum LPJZ-658, copper deficiency combined with a high-sugar diet-induced MASLD mouse model was utilized in this study. We fed 40-week-old (middle-aged) male C57BL/6 mice a copper-deficient and high-sugar diet for 16 weeks (CuDS), with supplementary LPJZ-658 for the last 6 weeks (CuDS + LPJZ-658). In this study, we measured body weight, liver weight, and serum biochemical markers. Lipid accumulation, histology, lipidomics, and sphingolipid metabolism-related enzyme expression were investigated to analyze liver function. Untargeted metabolomics was used to analyze the serum and the composition and abundance of intestinal flora. In addition, the correlation between differential liver lipid profiles, serum metabolites, and gut flora at the genus level was measured. The results show that LPJZ-658 significantly improves abnormal liver function and hepatic steatosis. The lipidomics analyses and metabolic pathway analysis identified sphingolipid, retinol, and glycerophospholipid metabolism as the most relevant metabolic pathways that characterized liver lipid dysregulation in the CuDS group. Consistently, RT-qPCR analyses revealed that the enzymes catalyzing sphingolipid metabolism that were significantly upregulated in the CuDS group were downregulated by the LPJZ-658 treatment. In addition, the serum metabolomics results indicated that the linoleic acid, taurine and hypotaurine, and ascorbate and aldarate metabolism pathways were associated with CuDS-induced MASLD. Notably, we found that treatment with LPJZ-658 partially reversed the changes in the differential serum metabolites. Finally, LPJZ-658 effectively regulated intestinal flora abnormalities and was significantly correlated with differential hepatic lipid species and serum metabolites. In conclusion, we elucidated the function and potential mechanisms of LPJZ-658 in alleviating copper deficiency combined with sugar-induced middle-aged MASLD and hope this will provide possible treatment strategies for improving MASLD.


Subject(s)
Copper , Liver , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Animals , Male , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Mice , Copper/blood , Liver/metabolism , Lipid Metabolism , Gastrointestinal Microbiome/drug effects , Disease Models, Animal , Probiotics/administration & dosage , Probiotics/pharmacology , Metabolomics , Lactobacillus plantarum , Lipidomics , Multiomics
11.
Nutrients ; 16(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38999780

ABSTRACT

Chronic inflammation is involved in the development of age-related diseases. Given its persistence, controlling chronic inflammation is essential for preventing age-related diseases. In this study, we investigated the effects of Enterococcus faecalis EC-12 (EC-12), which has immunomodulatory and antioxidant effects, on liver gene expression and aging phenomena in mice. Short-term EC-12 administration stimulated the expression of genes involved in lipid synthesis and metabolism in the liver. Furthermore, long-term EC-12 administration from 10 weeks to 1.5 years of age resulted in significant increases in blood interleukin (IL)-6 and IL-10 concentrations (both p < 0.05) and a significant decrease in the monocyte chemotactic protein-1 concentration (p < 0.05). These results indicated pathologic improvement, such as suppression of fat degeneration in the liver. These results suggest that continuous EC-12 intake from a young age can suppress liver function abnormalities, which is one of the aging phenomena in old age, and contribute to health in old age.


Subject(s)
Aging , Enterococcus faecalis , Liver , Animals , Liver/metabolism , Mice , Male , Interleukin-10/blood , Interleukin-10/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Chemokine CCL2/metabolism , Chemokine CCL2/blood , Probiotics/administration & dosage , Mice, Inbred C57BL , Lipid Metabolism
12.
Nutrients ; 16(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999802

ABSTRACT

Emerging evidence suggests that microbiota plays a crucial role in the development, progression, and therapeutic options in obesity and its comorbidities. This study assessed preoperative probiotic therapy's impact on bariatric treatment outcomes. A 12-week randomized, double-blind, placebo-controlled trial with 48 patients undergoing bariatric surgery was conducted. Participants received probiotics-Sanprobi Barrier-which contained nine strains of bacteria: Bifidobacterium bifidum W23, Bifidobacterium lactis W51 and W52, Lactobacillus acidophilus W37, Levilactobacillus brevis W63, Lacticaseibacillus casei W56, Ligilactobacillus salivarius W24, Lactococcus lactis W19, and Lactococcus lactis W58. Primary outcomes included excess body weight loss, body weight loss, and excess body mass index loss, with secondary objectives focusing on metabolic profiles. Surgical treatment of obesity significantly improved anthropometric and metabolic parameters. No significant differences were observed in primary outcomes or in secondary outcomes between groups at any time point post-surgery. Preoperative probiotics administration did not affect clinical outcomes 1, 3, or 6 months following bariatric surgery.


Subject(s)
Bariatric Surgery , Probiotics , Weight Loss , Humans , Probiotics/administration & dosage , Probiotics/therapeutic use , Double-Blind Method , Female , Male , Adult , Middle Aged , Obesity/surgery , Gastrointestinal Microbiome , Treatment Outcome , Preoperative Care/methods , Dietary Supplements , Body Weight
13.
Nutrients ; 16(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38999840

ABSTRACT

Inflammatory bowel diseases (IBDs), such as Crohn's disease (CD) and ulcerative colitis (UC), are chronic diseases of the digestive system with a multifactorial and not fully understood etiology. There is research suggesting that they may be initiated by genetic, immunological, and lifestyle factors. In turn, all of these factors play an important role in the modulation of intestinal microflora, and a significant proportion of IBD patients struggle with intestinal dysbiosis, which leads to the conclusion that intestinal microflora disorders may significantly increase the risk of developing IBD. Additionally, in IBD patients, Toll-like receptors (TLRs) produced by intestinal epithelial cells and dendritic cells treat intestinal bacterial antigens as pathogens, which causes a disruption of the immune response, resulting in the development of an inflammatory process. This may result in the occurrence of intestinal dysbiosis, which IBD patients are significantly vulnerable to. In this study, we reviewed scientific studies (in particular, systematic reviews with meta-analyses, being studies with the highest level of evidence) regarding the microflora of patients with IBD vs. the microflora in healthy people, and the use of various strains in IBD therapy.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/immunology , Crohn Disease/microbiology , Crohn Disease/immunology , Probiotics/therapeutic use , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/immunology
14.
Nutrients ; 16(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38999837

ABSTRACT

Introduction-Background: Data from experimental trials show that Crocus sativus L. (saffron) is considered to improve glycemia, lipid profile, and blood pressure and reduce oxidative stress. So far, clinical trials have been conducted in individuals with metabolic syndrome and Diabetes Mellitus type 2 (DMT-2). The purpose of this study is to assess the effectiveness of saffron in individuals with Diabetes Mellitus type 1 (DMT-1). PATIENTS-METHODS: 61 individuals with DMT-1, mean age 48 years old (48.3 ± 14.6), 26 females (42.6%) were randomized to receive a new oral supplement in sachets containing probiotics, prebiotics, magnesium, and Crocus sativus L. extract or placebo containing probiotics, prebiotics and magnesium daily for 6 months. Glycemic control was assessed with a continuous glucose monitoring system and laboratory measurement of HbA1c and lipid profile was also examined. Blood pressure at baseline and end of intervention was also measured. Individuals were either on a continuous subcutaneous insulin infusion with an insulin pump or in multiple daily injection regimens. Diabetes distress and satiety were assessed through a questionnaire and body composition was assessed with bioelectrical impedance. RESULTS: At the end of the intervention, the two groups differed significantly only in serum triglycerides (p = 0.049). After 6 months of treatment, a significant reduction in the active group was observed in glycated hemoglobin (p = 0.046) and serum triglycerides (p = 0.021) compared to baseline. The other primary endpoints (glycemic control, lipid profile, blood pressure) did not differ within the groups from baseline to end of intervention, and there was no significant difference between the two groups. Diabetes distress score improved significantly only in the active group (p = 0.044), suggesting an overall improvement in diabetes disease burden in these individuals but that was not significant enough between the two groups. CONCLUSIONS: A probiotic supplement with saffron extract improves serum triglycerides in well-controlled people with DMT-1 and may potentially be a valuable adjunct for enhancing glycemic control.


Subject(s)
Crocus , Diabetes Mellitus, Type 1 , Dietary Supplements , Plant Extracts , Humans , Crocus/chemistry , Female , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Male , Middle Aged , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/blood , Double-Blind Method , Adult , Blood Glucose/drug effects , Blood Glucose/metabolism , Glycated Hemoglobin/metabolism , Blood Pressure/drug effects , Glycemic Control/methods , Probiotics/administration & dosage , Lipids/blood
15.
Nutrients ; 16(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38999842

ABSTRACT

BACKGROUND: Probiotic supplementation in preterm neonates is standard practice in many centres across the globe. The impact of probiotic supplementation in the neonatal age group on the risk of hospitalisation in infancy has not been reported previously. METHODS: Infants born < 32 + 6 weeks of gestation in Western Australia were eligible for inclusion. We conducted a retrospective cohort study comparing data from before probiotic supplementation (Epoch 1: 1 December 2008-30 November 2010, n = 1238) versus after (Epoch 2: 1 June 2012-30 May 2014, n = 1422) on the risks of respiratory- and gastrointestinal infection-related hospitalisation. A subgroup analysis of infants born < 28 weeks of gestation was analysed separately for similar outcomes. RESULTS: Compared to Epoch 1, an 8% reduction in incidence of hospitalisation up to 2 years after birth was observed in Epoch 2 (adjusted incidence rate ratio (IRR) of 0.92; 95% confidence interval (CI); 0.87-0.98), adjusted for gestational age, smoking, socioeconomic status, and maternal age. The rate of hospitalisation for infants born < 28 weeks of gestation was comparable in epochs 1 and 2. CONCLUSION: Infants exposed to probiotic supplementation in the neonatal period experience a reduced risk of hospitalisation in the first two years after discharge from the neonatal unit.


Subject(s)
Dietary Supplements , Hospitalization , Probiotics , Humans , Western Australia/epidemiology , Infant, Newborn , Probiotics/administration & dosage , Probiotics/therapeutic use , Hospitalization/statistics & numerical data , Retrospective Studies , Female , Male , Infant , Gestational Age , Infant, Premature , Incidence , Risk Factors , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control
16.
Nutrients ; 16(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38999876

ABSTRACT

The increased global prevalence of chronic respiratory diseases in recent years has caused a substantial public health burden. Lactiplantibacillus plantarum KC3 and Leonurus japonicus Houtt. (LJH) extracts can alleviate respiratory symptoms and improve lung function in vitro and in vivo. However, the clinical efficacy and safety profile of this combination in patients with respiratory diseases remain unclear. Therefore, this multicenter, randomized, double-blind, placebo-controlled clinical trial aimed to evaluate the efficacy and safety of L. plantarum KC3 and LJH extracts in adults with respiratory discomfort. This mixture was termed 'CKDB-315'. Participants, randomly assigned to the CKDB-315 or placebo groups, were treated for 12 weeks. Assessments included the St. George's Respiratory Questionnaire (SGRQ) and the Chronic Obstructive Pulmonary Disease Assessment Test (CAT). The CKDB-315 group showed considerably improved SGRQ and CAT scores compared with the placebo group. Secondary outcomes, including dyspnea, pulmonary function, total antioxidant status, and inflammatory cytokine levels, were consistent with the primary outcomes. Exploratory analyses of the gut microbiota and short-chain fatty acid contents revealed the potential mechanisms underlying the effects of CKDB-315. Finally, safety analysis indicated that CKDB-315 was well tolerated and caused few adverse events. Our findings indicate that CKDB-315 is a promising therapeutic option for respiratory discomfort in adults.


Subject(s)
Leonurus , Plant Extracts , Probiotics , Humans , Double-Blind Method , Male , Female , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Middle Aged , Leonurus/chemistry , Probiotics/administration & dosage , Lactobacillus plantarum , Aged , Treatment Outcome , Gastrointestinal Microbiome/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/microbiology , Adult
17.
Nutrients ; 16(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38999886

ABSTRACT

Alcoholic liver damage is caused by long-term or heavy drinking, and it may further progress into alcoholic liver diseases (ALD). Probiotic supplements have been suggested for the prevention or improvement of liver damage. This study was designed to consider the ameliorative effects of Lactobacillus rhamnosus NKU FL1-8 isolated from infant feces against alcoholic liver damage. The mice were gavaged with a 50% ethanol solution and treated with 109 CFU of L. rhamnosus NKU FL1-8 suspension. The factors for liver function, oxidative stress, inflammation, gut microbiota composition, and intestinal barrier integrity were measured. The results showed that L. rhamnosus NKU FL1-8 could decrease the levels of aspartate aminotransferase (AST) to 61% and alanine aminotransferase (ALT) to 50% compared with ethanol given by gavage. It could inhibit the expression level of malondialdehyde (MDA), increase superoxide dismutase (SOD), glutathione (GSH) to relieve oxidative stress, and down-regulate the cytokines to decrease hepatic inflammation. After treatment, the level of triglycerides was reduced, and the expression levels of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and the peroxisome proliferators-activated receptor-α (PPAR-α) pathway were up-regulated. Additionally, the 16S rRNA sequencing analysis showed that L. rhamnosus NKU FL1-8 increased the relative abundance of Lactobacillus, Ruminococcaceae, etc. At the same time, L. rhamnosus NKU FL1-8 could significantly reduce lipopolysaccharides (LPS) and enhance intestinal tight junction proteins. These results demonstrated that L. rhamnosus NKU FL1-8 could reduce the level of oxidative stress, fat accumulation, and liver inflammation caused by alcohol in the host. The underlying mechanism could be that L. rhamnosus NKU FL1-8 inhibits LPS by regulating the gut microbiota and repairing the intestinal barrier. Thereby, these findings support L. rhamnosus NKU FL1-8 as a potential functional food for the relief of ALD.


Subject(s)
Feces , Gastrointestinal Microbiome , Lacticaseibacillus rhamnosus , Liver Diseases, Alcoholic , Mice, Inbred C57BL , Oxidative Stress , Probiotics , Animals , Gastrointestinal Microbiome/drug effects , Feces/microbiology , Oxidative Stress/drug effects , Liver Diseases, Alcoholic/prevention & control , Probiotics/pharmacology , Mice , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Male , Liver/drug effects , Liver/metabolism , Infant , Ethanol , Disease Models, Animal
18.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-39038938

ABSTRACT

With the increasing prevalence of age-related chronic diseases burdening healthcare systems, there is a pressing need for innovative management strategies. Our study focuses on the gut microbiota, essential for metabolic, nutritional, and immune functions, which undergoes significant changes with aging. These changes can impair intestinal function, leading to altered microbial diversity and composition that potentially influence health outcomes and disease progression. Using advanced metagenomic sequencing, we explore the potential of personalized probiotic supplements in 297 older adults by analyzing their gut microbiota. We identified distinctive Lactobacillus and Bifidobacterium signatures in the gut microbiota of older adults, revealing probiotic patterns associated with various population characteristics, microbial compositions, cognitive functions, and neuroimaging results. These insights suggest that tailored probiotic supplements, designed to match individual probiotic profile, could offer an innovative method for addressing age-related diseases and functional declines. Our findings enhance the existing evidence base for probiotic use among older adults, highlighting the opportunity to create more targeted and effective probiotic strategies. However, additional research is required to validate our results and further assess the impact of precision probiotics on aging populations. Future studies should employ longitudinal designs and larger cohorts to conclusively demonstrate the benefits of tailored probiotic treatments.


Subject(s)
Aging , Dietary Supplements , Gastrointestinal Microbiome , Probiotics , Probiotics/therapeutic use , Probiotics/administration & dosage , Humans , Aged , Female , Male , Aged, 80 and over , Middle Aged , Lactobacillus/genetics , Metagenomics/methods , Bifidobacterium
19.
BMC Microbiol ; 24(1): 253, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982403

ABSTRACT

BACKGROUND: Gut microbes play an important role in the growth and health of neonatal piglets. Probiotics can promote the healthy growth of neonatal piglets by regulating their gut microbes. The study investigated the effects of spraying Lactiplantibacillus plantarum P-8 (L. plantarum P-8) fermentation broth on the growth performance and gut microbes of neonatal piglets. RESULTS: The animals were randomly divided into probiotics groups (109 neonatal piglets) and control groups (113 neonatal piglets). The probiotics group was sprayed with L. plantarum P-8 fermented liquid from 3 day before the expected date of the sow to the 7-day-old of piglets, while the control group was sprayed with equal dose of PBS. Average daily gain (ADG), immune and antioxidant status and metagenome sequencing were used to assess the changes in growth performance and gut microbiota of neonatal piglets. The results showed that L. plantarum P-8 treatment significantly improved the average daily gain (P < 0.05) of neonatal piglets. L. plantarum P-8 increased the activities of CAT and SOD but reduced the levels of IL-2 and IL-6, effectively regulating the antioxidant capacity and immunity in neonatal piglets. L. plantarum P-8 adjusted the overall structure of gut microflora improving gut homeostasis to a certain extent, and significantly increased the relative abundance of gut beneficial bacteria such as L. mucosae and L. plantarum. CONCLUSION: Spraying L. plantarum P-8 can be a feasible and effective probiotic intervention not only improving the growth of neonatal piglets, regulating the antioxidant capacity and immunity of neonatal piglets, but also improving the gut homeostasis to a certain extent.


Subject(s)
Animals, Newborn , Gastrointestinal Microbiome , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Swine , Gastrointestinal Microbiome/drug effects , Lactobacillus plantarum , Fermentation , Antioxidants/metabolism , Antioxidants/administration & dosage , Antioxidants/pharmacology , Feces/microbiology
20.
Food Funct ; 15(14): 7658-7668, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38953736

ABSTRACT

Obesity is often accompanied by low-grade chronic inflammation and metabolic syndrome. It has been established that microbiota influences many physiological processes, including the development of obesity, and dysbiosis has been observed in obese individuals. In this study, we aimed to evaluate the impact of a new probiotic formulation, containing two probiotic strains and the bioactive compound octacosanol, on body weight, metabolic parameters, and concentrations of certain adipocytokines and appetite-regulating hormones in obese women. This double blind placebo-controlled supplementary intervention study included twenty-five women in the intervention group and twenty-three in the placebo group, and it lasted 12 weeks. Daily oral supplementation included 7 × 1010 CFU of Lactiplantibacillus plantarum 299v (DSM9843), 5 × 109 CFU of Saccharomyces cerevisiae var. boulardii (DBVPG6763), and 40 mg of octacosanol or placebo. Body weight, metabolic parameters, adipocytokines, and appetite-regulating hormones were assessed before (T0) and after the intervention (T1). After the intervention, significantly lower median concentrations of CRP (p = 0.005) and IL-6 (p = 0.012) were measured in the intervention group than the baseline, while the median concentrations of ghrelin (p = 0.026) and HDL-cholesterol (p = 0.03) were significantly increased. The intervention group had lower CRP levels (p = 0.023) and higher ghrelin levels (p = 0.006) than the placebo group. Significant changes in BMI between groups were not observed. In summary, although the new probiotic formulation showed beneficial effects on IL-6, CRP, HDL, and ghrelin levels, its potential effects on regulating triglyceride, insulin, and glucose levels require further studies before the novel dietary intervention could be considered a useful adjuvant therapy and an effective strategy for the management of obesity and obesity-associated comorbidities.


Subject(s)
Adipokines , Obesity , Probiotics , Humans , Female , Probiotics/pharmacology , Probiotics/therapeutic use , Obesity/diet therapy , Obesity/metabolism , Double-Blind Method , Adult , Adipokines/blood , Adipokines/metabolism , Middle Aged , Ghrelin/blood , Appetite/drug effects , Lactobacillus plantarum , Body Weight/drug effects , C-Reactive Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL