Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 620
Filter
Add more filters

Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G712-G725, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38626403

ABSTRACT

Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract , Humans , Animals , Gastrointestinal Tract/immunology , Gastrointestinal Tract/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Microbiome/physiology , Neuroimmunomodulation/physiology , Enteric Nervous System/physiology , Enteric Nervous System/immunology
2.
J Sci Food Agric ; 104(11): 6359-6375, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38334314

ABSTRACT

Dietary patterns play an important role in regards to the modulation and control of the gut microbiome composition and function. The interaction between diet and microbiota plays an important role in order to maintain intestinal homeostasis, which ultimately affect the host's health. Diet directly impacts the microbes that inhabit the gastrointestinal tract (GIT), which then contributes to the production of secondary metabolites, such as short-chain fatty acids, neurotransmitters, and antimicrobial peptides. Dietary consumption with genetically modified probiotics can be the best vaccine delivery vector and protect cells from various illnesses. A holistic approach to disease prevention, treatment, and management takes these intrinsically linked diet-microbes, microbe-microbe interactions, and microbe-host interactions into account. Dietary components, such as fiber can modulate beneficial gut microbiota, and they have resulting ameliorative effects against metabolic disorders. Medical interventions, such as antibiotic drugs can conversely have detrimental effects on gut microbiota by disputing the balance between Bacteroides and firmicute, which contribute to continuing disease states. We summarize the known effects of various dietary components, such as fibers, carbohydrates, fatty acids, vitamins, minerals, proteins, phenolic acids, and antibiotics on the composition of the gut microbiota in this article in addition to the beneficial effect of genetically modified probiotics and consequentially their role in regards to shaping human health. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Probiotics/administration & dosage , Humans , Animals , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Diet
3.
Cell Host Microbe ; 32(3): 396-410.e6, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38359828

ABSTRACT

Antibiotic resistance and evasion are incompletely understood and complicated by the fact that murine interval dosing models do not fully recapitulate antibiotic pharmacokinetics in humans. To better understand how gastrointestinal bacteria respond to antibiotics, we colonized germ-free mice with a pan-susceptible genetically barcoded Escherichia coli clinical isolate and administered the antibiotic cefepime via programmable subcutaneous pumps, allowing closer emulation of human parenteral antibiotic dynamics. E. coli was only recovered from intestinal tissue, where cefepime concentrations were still inhibitory. Strikingly, "some" E. coli isolates were not cefepime resistant but acquired mutations in genes involved in polysaccharide capsular synthesis increasing their invasion and survival within human intestinal cells. Deleting wbaP involved in capsular polysaccharide synthesis mimicked this phenotype, allowing increased invasion of colonocytes where cefepime concentrations were reduced. Additionally, "some" mutant strains exhibited a persister phenotype upon further cefepime exposure. This work uncovers a mechanism allowing "select" gastrointestinal bacteria to evade antibiotic treatment.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Humans , Animals , Mice , Cefepime , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Gastrointestinal Tract/microbiology , Polysaccharides , Microbial Sensitivity Tests , Mammals
4.
Microb Biotechnol ; 16(1): 34-53, 2023 01.
Article in English | MEDLINE | ID: mdl-36415969

ABSTRACT

The effects of plastic debris on the environment and plant, animal, and human health are a global challenge, with micro(nano)plastics (MNPs) being the main focus. MNPs are found so often in the food chain that they are provoking an increase in human intake. They have been detected in most categories of consumed foods, drinking water, and even human feces. Therefore, oral ingestion becomes the main source of exposure to MNPs, and the gastrointestinal tract, primarily the gut, constantly interacts with these small particles. The consequences of human exposure to MNPs remain unclear. However, current in vivo studies and in vitro gastrointestinal tract models have shown that MNPs of several types and sizes impact gut intestinal bacteria, affecting gut homeostasis. The typical microbiome signature of MNP ingestion is often associated with dysbiosis and loss of resilience, leads to frequent pathogen outbreaks, and local and systemic metabolic disorders. Moreover, the small micro- and nano-plastic particles found in animal tissues with accumulated evidence of microbial degradation of plastics/MNPs by bacteria and insect gut microbiota raise the issue of whether human gut bacteria make key contributions to the bio-transformation of ingested MNPs. Here, we discuss these issues and unveil the complex interplay between MNPs and the human gut microbiome. Therefore, the elucidation of the biological consequences of this interaction on both host and microbiota is undoubtedly challenging. It is expected that microbial biotechnology and microbiome research could help decipher the extent to which gut microorganisms diversify and MNP-determinant species, mechanisms, and enzymatic systems, as well as become important to understand our response to MNP exposure and provide background information to inspire future holistic studies.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Humans , Gastrointestinal Tract/microbiology , Bacteria/genetics , Feces/microbiology
5.
Nutrients ; 14(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35057561

ABSTRACT

Parkinson's disease is characterized by motor and non-motor symptoms, such as defects in the gut function, which may occur before the motor symptoms. To date, there are therapies that can improve these symptoms, but there is no cure to avoid the development or exacerbation of this disorder. Dysbiosis of gut microbiota could have a crucial role in the gut-brain axis, which is a bidirectional communication between the central nervous system and the enteric nervous system. Diet can affect the microbiota composition, impacting gut-brain axis functionality. Gut microbiome restoration through probiotics, prebiotics, synbiotics or other dietary means could have the potential to slow PD progression. In this review, we will discuss the influence of diet on the bidirectional communication between gut and brain, thus supporting the hypothesis that this disorder could begin in the gut. We also focus on how food-based therapies might then have an influence on PD and could ameliorate non-motor as well as motor symptoms.


Subject(s)
Brain-Gut Axis/physiology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Diet , Disease Progression , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Humans , Nutrition Therapy , Prebiotics/administration & dosage , Probiotics/therapeutic use , Synbiotics/administration & dosage
6.
Int J Med Microbiol ; 312(2): 151548, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35030401

ABSTRACT

BACKGROUND: Three healthy volunteers carried similar quinolone-resistant E. coli (QREC) (pulsed field gel electrophoresis profiles) in their gut before and after 14 days ciprofloxacin treatment. Given the intensity of the selective pressure and the mutagenic properties of quinolones, we determined whether these strains had evolved at the phenotypic and/or genomic levels. MATERIAL AND METHODS: Commensal QREC from before day-0 (D0), and a month after 14 days of ciprofloxacin (D42) were compared in 3 volunteers. Growth experiments were performed; acetate levels, mutation frequencies, quinolone MICs and antibiotic tolerance were measured at D0 and D42. Genomes were sequenced and single nucleotide polymorphisms (SNPs) between D0 and D42 were analyzed using DiscoSNP and breseq methods. Cytoplasmic proteins were extracted, HPLC performed and proteins identified using X!tandem software; abundances were measured by mass spectrometry using the Spectral Counting (SC) and eXtraction Ion Chromatograms (XIC) integration methods. RESULTS: No difference was found in MICs, growth characteristics, acetate concentrations, mutation frequencies, tolerance profiles, phylogroups, O-and H-types, fimH alleles and sequence types between D0 and D42. No SNP variation was evidenced between D0 and D42 isolates for 2/3 subjects; 2 SNP variations were evidenced in one. At the protein level, very few significant protein abundance differences were identified between D0 and D42. CONCLUSION: No fitness, tolerance, metabolic or genomic evolution of commensal QREC was observed overtime, despite massive exposure to ciprofloxacin in the gut. The three strains behaved as if they had been unaffected by ciprofloxacin, suggesting that gut may act as a sanctuary where bacteria would be protected from the effect of antibiotics and survive without any detrimental effect of stress.


Subject(s)
Ciprofloxacin , Escherichia coli Infections , Escherichia coli , Gastrointestinal Tract/microbiology , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Fluoroquinolones/pharmacology , Humans , Microbial Sensitivity Tests
7.
J Nutr Biochem ; 100: 108904, 2022 02.
Article in English | MEDLINE | ID: mdl-34748918

ABSTRACT

Neuroinflammation is a central factor in neuropathic pain (NP). Ginger is a promising bioactive compound in NP management due to its anti-inflammatory property. Emerging evidence suggests that gut microbiome and gut-derived metabolites play a key role in NP. We evaluated the effects of two ginger root extracts rich in gingerols (GEG) and shogaols (SEG) on pain sensitivity, anxiety-like behaviors, circulating cell-free mitochondrial DNA (ccf-mtDNA), gut microbiome composition, and fecal metabolites in rats with NP. Sixteen male rats were divided into four groups: sham, spinal nerve ligation (SNL), SNL+0.75%GEG in diet, and SNL+0.75%SEG in diet groups for 30 days. Compared to SNL group, both SNL+GEG and SNL+SEG groups showed a significant reduction in pain- and anxiety-like behaviors, and ccf-mtDNA level. Relative to the SNL group, both SNL+GEG and SNL+SEG groups increased the relative abundance of Lactococcus, Sellimonas, Blautia, Erysipelatoclostridiaceae, and Anaerovoracaceae, but decreased that of Prevotellaceae UCG-001, Rikenellaceae RC9 gut group, Mucispirillum and Desulfovibrio, Desulfovibrio, Anaerofilum, Eubacterium siraeum group, RF39, UCG-005, Lachnospiraceae NK4A136 group, Acetatifactor, Eubacterium ruminantium group, Clostridia UCG-014, and an uncultured Anaerovoracaceae. GEG and SEG had differential effects on gut-derived metabolites. Compared to SNL group, SNL+GEG group had higher level of 1'-acetoxychavicol acetate, (4E)-1,7-Bis(4-hydroxyphenyl)-4-hepten-3-one, NP-000629, 7,8-Dimethoxy-3-(2-methyl-3-buten-2-yl)-2H-chromen-2-one, 3-{[4-(2-Pyrimidinyl)piperazino]carbonyl}-2-pyrazinecarboxylic acid, 920863, and (1R,3R,7R,13S)-13-Methyl-6-methylene-4,14,16-trioxatetracyclo[11.2.1.0∼1,10∼.0∼3,7∼]hexadec-9-en-5-one, while SNL+SEG group had higher level for (±)-5-[(tert-Butylamino)-2'-hydroxypropoxy]-1_2_3_4-tetrahydro-1-naphthol and dehydroepiandrosteronesulfate. In conclusion, ginger is a promising functional food in the management of NP, and further investigations are necessary to assess the role of ginger on gut-brain axis in pain management.


Subject(s)
Bacteria/metabolism , Catechols/administration & dosage , Dietary Supplements , Fatty Alcohols/administration & dosage , Gastrointestinal Microbiome , Neuralgia/diet therapy , Plant Extracts , Zingiber officinale , Animals , DNA, Mitochondrial/blood , Feces/chemistry , Gastrointestinal Tract/microbiology , Ligation , Male , Pain Management , Rats , Rats, Sprague-Dawley , Spinal Nerves
8.
J Nutr Biochem ; 101: 108926, 2022 03.
Article in English | MEDLINE | ID: mdl-34848335

ABSTRACT

Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.


Subject(s)
Butyrates/administration & dosage , Dietary Supplements , Fatty Acids, Volatile/administration & dosage , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Animals , Cell Differentiation , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colon/pathology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
9.
Sci Rep ; 11(1): 24373, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934118

ABSTRACT

Gestational Diabetes Mellitus (GDM) and obesity affect the functioning of multiple maternal systems and influence colonization of the newborn gastrointestinal through the breastmilk microbiota (BMM). It is currently unclear how GDM and obesity affect the human BMM composition. Here, we applied 16S-rRNA high-throughput sequencing to human colostrum milk to characterize BMM taxonomic changes in a cohort of 43 individuals classified in six subgroups according to mothers patho-physiological conditions (healthy control (n = 18), GDM (n = 13), or obesity (n = 12)) and newborn gender. Using various diversity indicators, including Shannon/Faith phylogenetic index and UniFrac/robust Aitchison distances, we evidenced that BMM composition was influenced by the infant gender in the obesity subgroup. In addition, the GDM group presented higher microbial diversity compared to the control group. Staphylococcus, Corynebacterium 1, Anaerococcus and Prevotella were overrepresented in colostrum from women with either obesity or GDM, compared to control samples. Finally, Rhodobacteraceae was distinct for GDM and 5 families (Bdellovibrionaceae, Halomonadaceae, Shewanellaceae, Saccharimonadales and Vibrionaceae) were distinct for obesity subgroups with an absolute effect size greater than 1 and a q-value ≤ 0.05. This study represents the first effort to describe the impact of maternal GDM and obesity on BMM.


Subject(s)
Bacteria/genetics , Colostrum/microbiology , Diabetes, Gestational/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Milk, Human/microbiology , Obesity/microbiology , Adult , Bacteria/classification , Bacteria/isolation & purification , Body Mass Index , Female , Humans , Infant, Newborn , Male , Phylogeny , Pregnancy
10.
PLoS One ; 16(12): e0261189, 2021.
Article in English | MEDLINE | ID: mdl-34941893

ABSTRACT

Because of a possible impact of capsaicin in the high concentrations on enterocyte injury (cytotoxicity) and bactericidal activity on probiotics, Lactobacillus rhamnosus L34 (L34) and Lactobacillus rhamnosus GG (LGG), the probiotics derived from Thai and Caucasian population, respectively, were tested in the chili-extract administered C57BL/6 mice and in vitro experiments. In comparison with placebo, 2 weeks administration of the extract from Thai chili in mice caused loose feces and induced intestinal permeability defect as indicated by FITC-dextran assay and the reduction in tight junction molecules (occludin and zona occludens-1) using fluorescent staining and gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the chili extracts also induced the translocation of gut pathogen molecules; lipopolysaccharide (LPS) and (1→3)-ß-d-glucan (BG) and fecal dysbiosis (microbiome analysis), including reduced Firmicutes, increased Bacteroides, and enhanced total Gram-negative bacteria in feces. Both L34 and LGG attenuated gut barrier defect (FITC-dextran, the fluorescent staining and gene expression of tight junction molecules) but not improved fecal consistency. Additionally, high concentrations of capsaicin (0.02-2 mM) damage enterocytes (Caco-2 and HT-29) as indicated by cell viability test, supernatant cytokine (IL-8), transepithelial electrical resistance (TEER) and transepithelial FITC-dextran (4.4 kDa) but were attenuated by Lactobacillus condition media (LCM) from both probiotic-strains. The 24 h incubation with 2 mM capsaicin (but not the lower concentrations) reduced the abundance of LGG (but not L34) implying a higher capsaicin tolerance of L34. However, Lactobacillus rhamnosus fecal abundance, using qRT-PCR, of L34 or LGG after 3, 7, and 20 days of the administration in the Thai healthy volunteers demonstrated the similarity between both strains. In conclusion, high dose chili extracts impaired gut permeability and induced gut dysbiosis but were attenuated by probiotics. Despite a better capsaicin tolerance of L34 compared with LGG in vitro, L34 abundance in feces was not different to LGG in the healthy volunteers. More studies on probiotics with a higher intake of chili in human are interesting.


Subject(s)
Capsaicin/adverse effects , Dysbiosis/prevention & control , Gastrointestinal Tract/drug effects , Inflammation/prevention & control , Lacticaseibacillus rhamnosus/chemistry , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Anti-Bacterial Agents/administration & dosage , Antipruritics/administration & dosage , Antipruritics/adverse effects , Capsaicin/administration & dosage , Cytokines/metabolism , Dysbiosis/chemically induced , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Humans , Inflammation/chemically induced , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Plant Extracts/administration & dosage , Plant Extracts/adverse effects , Probiotics/adverse effects , Tight Junctions , Young Adult
11.
Nutrients ; 13(11)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34836186

ABSTRACT

We studied whether probiotics were beneficial for hormonal change-associated dysbiosis, which may influence the enteric nervous system and GI function during early pregnancy. The study was 16 days consisting of two cycles of six daily probiotics mainly Lactobacillus and 2 days without probiotics. Daily surveys were conducted to monitor GI function and life quality. A subset of the participants who contributed fecal specimens was used for microbiota metagenomic sequencing, metabolomics, and quantification of bacterial genes to understand potential underlying mechanisms. Statistical analyses were done by generalized linear mixed-effects models. Thirty-two obstetric patients and 535 daily observations were included. The data revealed that probiotic supplementation significantly reduced the severity of nausea, vomiting, constipation, and improved life quality. Moreover, a low copy number of fecal bsh (bile salt hydrolase), which generates free bile acids, was associated with high vomiting scores and probiotic intake increased fecal bsh. In exploratory analysis without adjusting for multiplicity, a low fecal α-tocopherol, as well as a high abundance of Akkemansia muciniphila, was associated with high vomiting scores and times, respectively. The potential implications of these biomarkers in pregnancy and GI function are discussed. Probiotics likely produce free bile acids to facilitate intestinal mobility and metabolism.


Subject(s)
Dysbiosis/drug therapy , Gastrointestinal Tract/metabolism , Pregnancy Complications/drug therapy , Probiotics/administration & dosage , Quality of Life , Adult , Akkermansia , Amidohydrolases/metabolism , Bile Acids and Salts/metabolism , Feces/microbiology , Female , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Humans , Lactobacillus , Metabolomics/methods , Nausea/drug therapy , Pregnancy , Vomiting/drug therapy
12.
Crit Rev Microbiol ; 47(5): 596-611, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34407384

ABSTRACT

The human gastrointestinal (GI) tract hosts trillions of microbial inhabitants involved in maintaining intestinal homeostasis, dysbiosis of which provokes a motley of pathogenic and autoimmune disorders. While the mechanisms by which the microbiota modulates human health are manifold, their liberated metabolites from ingested dietary supplements play a crucial role by bidirectionally regulating the expression of micro-ribonucleic acids (miRNAs). miRNAs are small endogenous non-coding RNAs (ncRNAs) that have been confirmed to be involved in an interplay with microbiota to regulate host gene expression. This comprehensive review focuses on key principles of miRNAs, their regulation, and crosstalk with gut microbiota to influence host gene expression in various human disorders, by bringing together important recent findings centric around miRNA-microbiota interactions in diseases along various axis of the gut with other organs. We also attempt to lay emphasis on exploiting the avenues of gut-directed miRNA therapeutics using rudimentary dietary supplements to regulate abnormal host gene expression in diseases, opening doors to an accessible and economical therapeutic strategy.


Subject(s)
Gastrointestinal Microbiome , Gene Expression Regulation , MicroRNAs/genetics , Therapeutics , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/microbiology , Cardiovascular Diseases/therapy , Digestive System Diseases/genetics , Digestive System Diseases/microbiology , Digestive System Diseases/therapy , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Health , Humans , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/microbiology , Neoplasms/therapy , Nervous System Diseases/genetics , Nervous System Diseases/microbiology , Nervous System Diseases/therapy
13.
Acta Biochim Pol ; 68(3): 393-398, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34432408

ABSTRACT

The time of COVID-19 pandemic focused the attention of scientist to recognise the complex medical symptoms of the disease, modes of infection and possible therapies. The organisms' response towards SARS-CoV-2 infection depends on many individual factors and the course of disease is described as unprecedented and complex. Numerous symptoms from the respiratory system, abnormalities in the gastrointestinal tract, stroke, liver damage and coagulopathy, among others, are accompanied by negative side effects of the pandemic lifestyle, including immunity depletion, overall fitness impairment, skin condition worsening, psychological and psychiatric consequences. There is an urgent need to seek all possible routes for assuring favouring conditions to build and support the organisms' microbiological barriers and enhance immunity, which will also help during the ongoing vaccination action. Probiotic Lactic Acid Bacteria (LAB) and environmental Bacillus species are microorganisms typically found in food products or dietary supplements, but also applied on body surfaces or technological surfaces at home and in the industry. Since the contemporary definition of probiotics points to positive health effects, it is of highest importance to follow strict regulations and standards of product manufacturing, especially in the times of biohazard risks and rising public distrust of therapies. There is an urgent need to seek all possible routes for assuring the favouring conditions to build and support the organisms' microbiological barriers and enhance the immunity, that will serve also during the ongoing vaccination action. Probiotic LAB and environmental Bacillus species are microorganisms typically found in food products or dietary supplements, but also applied on body surface or technological surfaces in household and industry. Since the contemporary definition of probiotics points out the positive health effects, it is of highest importance to follow strict regulations and standards of product manufacturing, especially in the times of biohazard and rising public distrust of therapies.


Subject(s)
COVID-19/immunology , Probiotics/therapeutic use , COVID-19/microbiology , COVID-19/virology , Dietary Supplements , Gastrointestinal Tract/microbiology , Humans , SARS-CoV-2/isolation & purification
14.
Nutrients ; 13(8)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34444706

ABSTRACT

Cranberry is a fruit originally from New England and currently growing throughout the east and northeast parts of the USA and Canada. The supplementation of cranberry extracts as nutraceuticals showed to contribute to the prevention of urinary tract infections, and most likely it may help to prevent cardiovascular and gastroenteric diseases, as highlighted by several clinical trials. However, aiming to validate the efficacy and safety of clinical applications as long-term randomized clinical trials (RCTs), further investigations of the mechanisms of action are required. In addition, a real challenge for next years is the standardization of cranberry's polyphenolic fractions. In this context, the optimization of the extraction process and downstream processing represent a key point for a reliable active principle for the formulation of a food supplement. For this reason, new non-conventional extraction methods have been developed to improve the quality of the extracts and reduce the overall costs. The aim of this survey is to describe both technologies and processes for highly active cranberry extracts as well as the effects observed in clinical studies and the respective tolerability notes.


Subject(s)
Dietary Supplements , Phytochemicals , Plant Extracts , Vaccinium macrocarpon , Animals , Blood Glucose/metabolism , Female , Food Handling , Fruit , Fruit and Vegetable Juices , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Helicobacter Infections/diet therapy , Helicobacter pylori , Humans , Inflammation/prevention & control , Insulin/blood , Male , Phytochemicals/analysis , Phytochemicals/isolation & purification , Plant Extracts/isolation & purification , Polyphenols , Urinary Tract Infections/diet therapy , Urinary Tract Infections/prevention & control , Vaccinium macrocarpon/chemistry
15.
Mol Biol Rep ; 48(6): 5259-5273, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34228274

ABSTRACT

BACKGROUND: Dysfunction of the gastrointestinal tract (GIT) is one of the most common non-motor symptom of Parkinson's Disease (PD). Pathological processes causing PD were suggested to initiate in the enteric nervous system (ENS) and proceed to the central nervous system (CNS). There are studies showing that low-carbohydrate ketogenic diets can improve motor symptoms of PD. Caprylic acid (C8) is the principal fatty acid component of the medium-chain triglycerides in the ketogenic diets. In this study, we aimed to evaluate the effects of caprylic acid, in neurotoxin exposed zebrafish focusing on the relationship between intestinal and brain oxidative stress and inflammation. METHODS: Adult zebrafish were exposed to rotenone (5 µg/L) (R group) and caprylic acid (20 and 60 mg/mL) (L + HDCA and R + HDCA groups) for 30 days. At the end of 30 days locomotor activities were determined. Levels of lipid peroxidation (LPO), nitric oxide, glutathione and superoxide dismutase and glutathione S-transferase activities were determined by spectrophotometric methods and gene expressions of tnf⍺, il1, il6, il21, ifnÉ£ and bdnf were evaluated by RT-PCR in the brain and intestinal tissues of zebrafish. RESULTS: Caprylic acid ameliorated LPO, NO, SOD and the expressions of tnf⍺, il1, il6, il21, ifnÉ£ and bdnf in brain and intestines. Locomotor activities were only ameliorated in high dose R + HDCA group. CONCLUSIONS: Caprylic acid ameliorated the neurotoxin-induced oxidative stress and inflammation both in the brain and intestines and enhanced locomotor activity in zebrafish.


Subject(s)
Brain-Gut Axis/physiology , Caprylates/pharmacology , Animals , Brain/metabolism , Brain-Gut Axis/drug effects , Caprylates/metabolism , Disease Models, Animal , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Glutathione/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Rotenone/adverse effects , Superoxide Dismutase/metabolism , Zebrafish , Zebrafish Proteins
16.
Food Funct ; 12(14): 6157-6166, 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34079965

ABSTRACT

The intra-amniotic administration approach has been used to evaluate the effects of plant origin prebiotics on intestinal health and on brush border membrane functionality and morphology. Prebiotics are fermentable dietary fibers, which can positively affect the host by selectively stimulating the growth and activity of colon bacteria, thus improving intestinal health. The consumption of prebiotics increases digestive tract motility, which leads to hyperplasia and/or hypertrophy of intestinal cells, increasing nutrient digestive and absorptive surface area. This review collates information about the effects and relationship between prebiotic consumption on small intestinal brush border membrane functionality and morphology by utilizing the intra-amniotic administration approach. To date, research has shown that the intra-amniotic administration of prebiotics affects the expression of key brush border membrane functional proteins, intestinal surface area (villi height/width), and goblet cell number/size. These effects may improve brush border membrane functionality and digestive/absorptive capabilities.


Subject(s)
Chickens , Intestinal Mucosa/drug effects , Microvilli/drug effects , Plant Extracts/pharmacology , Prebiotics/administration & dosage , Animals , Colon/microbiology , Dietary Fiber/administration & dosage , Digestion , Duodenum/metabolism , Duodenum/microbiology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Motility , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestines/microbiology , Microvilli/metabolism
17.
Cells ; 10(3)2021 03 22.
Article in English | MEDLINE | ID: mdl-33809924

ABSTRACT

Numerous honeybee (Apis mellifera) products, such as honey, propolis, and bee venom, are used in traditional medicine to prevent illness and promote healing. Therefore, this insect has a huge impact on humans' way of life and the environment. While the population of A. mellifera is large, there is concern that widespread commercialization of beekeeping, combined with environmental pollution and the action of bee pathogens, has caused significant problems for the health of honeybee populations. One of the strategies to preserve the welfare of honeybees is to better understand and protect their natural microbiota. This paper provides a unique overview of the latest research on the features and functioning of A. mellifera. Honeybee microbiome analysis focuses on both the function and numerous factors affecting it. In addition, we present the characteristics of lactic acid bacteria (LAB) as an important part of the gut community and their special beneficial activities for honeybee health. The idea of probiotics for honeybees as a promising tool to improve their health is widely discussed. Knowledge of the natural gut microbiota provides an opportunity to create a broad strategy for honeybee vitality, including the development of modern probiotic preparations to use instead of conventional antibiotics, environmentally friendly biocides, and biological control agents.


Subject(s)
Bees/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Honey , Lactobacillales/metabolism , Animals , Beekeeping , Bees/drug effects , Bees/metabolism , Dysbiosis , Host-Pathogen Interactions , Humans , Insecticides/adverse effects , Pest Control, Biological , Probiotics
18.
Am J Chin Med ; 49(3): 609-626, 2021.
Article in English | MEDLINE | ID: mdl-33683187

ABSTRACT

Cancer is a disease with a high mortality and disability rate. Cancer consists not only of cancer cells, but also of the surrounding microenvironment and tumor microenvironment (TME) constantly interacting with tumor cells to support tumor development and progression. Over the last decade, accumulating evidence has implicated that microbiota profoundly influences cancer initiation and progression. Most research focuses on gut microbiota, for the gut harbors the largest collection of microorganisms. Gut microbiota includes bacteria, viruses, protozoa, archaea, and fungi in the gastrointestinal tract, affecting DNA damage, host immune response and chronic inflammation in various types of cancer (i.e., colon cancer, gastric cancer and breast cancer). Notably, gut dysbiosis can reshape tumor microenvironment and make it favorable for tumor growth. Recently, accumulating studies have attached the importance of traditional Chinese medicine (TCM) to cancer treatments, and the bioactive natural compounds have been considered as potential drug candidates to suppress cancer initiation and development. Interestingly, more recent studies demonstrate that TCM could potentially prevent and suppress early-stage cancer progression through the regulation of gut microbiota. This review is on the purpose of exhausting the significance of gut microbiota in the tumor microenvironment as potential targets of Chinese medicine.


Subject(s)
Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Medicine, Chinese Traditional , Neoplasms/microbiology , Neoplasms/pathology , Tumor Microenvironment/physiology , DNA Damage , Disease Progression , Host-Pathogen Interactions/immunology , Humans , Neoplasms/drug therapy , Neoplasms/prevention & control , Phytotherapy
19.
Nat Commun ; 12(1): 942, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574256

ABSTRACT

The mosquito microbiota impacts the physiology of its host and is essential for normal larval development, thereby influencing transmission of vector-borne pathogens. Germ-free mosquitoes generated with current methods show larval stunting and developmental deficits. Therefore, functional studies of the mosquito microbiota have so far mostly been limited to antibiotic treatments of emerging adults. In this study, we introduce a method to produce germ-free Aedes aegypti mosquitoes. It is based on reversible colonisation with bacteria genetically modified to allow complete decolonisation at any developmental stage. We show that, unlike germ-free mosquitoes previously produced using sterile diets, reversibly colonised mosquitoes show no developmental retardation and reach the same size as control adults. This allows us to uncouple the study of the microbiota in larvae and adults. In adults, we detect no impact of bacterial colonisation on mosquito fecundity or longevity. In larvae, data from our transcriptome analysis and diet supplementation experiments following decolonisation suggest that bacteria support larval development by contributing to folate biosynthesis and by enhancing energy storage. Our study establishes a tool to study the microbiota in insects and deepens our knowledge on the metabolic contribution of bacteria to mosquito development.


Subject(s)
Host Microbial Interactions/physiology , Microbiota/physiology , Mosquito Vectors/microbiology , Aedes/genetics , Aedes/growth & development , Aedes/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Folic Acid , Food, Fortified , Gastrointestinal Tract/microbiology , Gene Expression Regulation , Germ-Free Life , Larva/genetics , Larva/growth & development , Larva/microbiology , Lipid Metabolism , Mosquito Vectors/growth & development , RNA, Ribosomal, 16S
20.
J Sci Food Agric ; 101(12): 5049-5055, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33570782

ABSTRACT

BACKGROUND: Previous studies have demonstrated the acrylamide-removing properties of probiotic monocultures; however, potential advantages of consortia over monocultures in reducing the dietary exposure to acrylamide have not been proven. Hence this work aims to assess the acrylamide (AA)-binding properties of bacterial consortia, consisting of either probiotic strains and / or representative bacteria of duodenal microbiota, exposed to simulated gastrointestinal conditions (SGC). The AA binding capacity of ten probiotic strains (PS) and six duodenal strains (NDS) was evaluated under different conditions; then, three different consortia (PS, NDS, and PS + NDS) were assessed under SGC. RESULTS: Among individual PS, Bacillus coagulans GBI-30, Lactobacillus fermentum J23, L. pentosus J37 and J24, and L. casei Shirota, exhibited the highest AA-binding capacity (80-87%), while Bifidobacterium catenulatun ATCC27676, Streptococcus salivarius subsp. thermophilus ATCC19258, and S. gallolyticus ATCC9809 were the best (ca. 68%) NDS monocultures. Probiotic strain consortia showed higher (P < 0.05) AA binding capacity (> 90%) than monoculture bacteria. Conversely, individual NDS cultures displayed higher (P < 0.05) binding capacity than NDS consortia (60%). A significant reduction (P < 0.05) in AA removal capacity was observed when consortia were exposed to SGC, PS consortia being the most effective (> 60% removal). CONCLUSION: These results suggest that consortia of specific PS could play an important role in reducing the intestinal availability of acrylamide. © 2021 Society of Chemical Industry.


Subject(s)
Acrylamide/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/microbiology , Lactobacillus/metabolism , Probiotics/pharmacology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Drug Evaluation, Preclinical , Gastrointestinal Tract/metabolism , Humans , Lactobacillus/classification , Lactobacillus/growth & development , Microbial Viability , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL