Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
Add more filters

Publication year range
1.
Cell ; 176(3): 649-662.e20, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30661755

ABSTRACT

The body-wide human microbiome plays a role in health, but its full diversity remains uncharacterized, particularly outside of the gut and in international populations. We leveraged 9,428 metagenomes to reconstruct 154,723 microbial genomes (45% of high quality) spanning body sites, ages, countries, and lifestyles. We recapitulated 4,930 species-level genome bins (SGBs), 77% without genomes in public repositories (unknown SGBs [uSGBs]). uSGBs are prevalent (in 93% of well-assembled samples), expand underrepresented phyla, and are enriched in non-Westernized populations (40% of the total SGBs). We annotated 2.85 M genes in SGBs, many associated with conditions including infant development (94,000) or Westernization (106,000). SGBs and uSGBs permit deeper microbiome analyses and increase the average mappability of metagenomic reads from 67.76% to 87.51% in the gut (median 94.26%) and 65.14% to 82.34% in the mouth. We thus identify thousands of microbial genomes from yet-to-be-named species, expand the pangenomes of human-associated microbes, and allow better exploitation of metagenomic technologies.


Subject(s)
Metagenome/genetics , Metagenomics/methods , Microbiota/genetics , Big Data , Genetic Variation/genetics , Geography , Humans , Life Style , Phylogeny , Sequence Analysis, DNA/methods
2.
Nature ; 614(7946): 125-135, 2023 02.
Article in English | MEDLINE | ID: mdl-36653448

ABSTRACT

The human microbiome is an integral component of the human body and a co-determinant of several health conditions1,2. However, the extent to which interpersonal relations shape the individual genetic makeup of the microbiome and its transmission within and across populations remains largely unknown3,4. Here, capitalizing on more than 9,700 human metagenomes and computational strain-level profiling, we detected extensive bacterial strain sharing across individuals (more than 10 million instances) with distinct mother-to-infant, intra-household and intra-population transmission patterns. Mother-to-infant gut microbiome transmission was considerable and stable during infancy (around 50% of the same strains among shared species (strain-sharing rate)) and remained detectable at older ages. By contrast, the transmission of the oral microbiome occurred largely horizontally and was enhanced by the duration of cohabitation. There was substantial strain sharing among cohabiting individuals, with 12% and 32% median strain-sharing rates for the gut and oral microbiomes, and time since cohabitation affected strain sharing more than age or genetics did. Bacterial strain sharing additionally recapitulated host population structures better than species-level profiles did. Finally, distinct taxa appeared as efficient spreaders across transmission modes and were associated with different predicted bacterial phenotypes linked with out-of-host survival capabilities. The extent of microorganism transmission that we describe underscores its relevance in human microbiome studies5, especially those on non-infectious, microbiome-associated diseases.


Subject(s)
Bacteria , Disease Transmission, Infectious , Gastrointestinal Microbiome , Home Environment , Microbiota , Mouth , Female , Humans , Infant , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Gastrointestinal Microbiome/genetics , Metagenome , Microbiota/genetics , Mothers , Mouth/microbiology , Infectious Disease Transmission, Vertical , Family Characteristics , Aging , Time Factors , Microbial Viability
3.
Environ Res ; 257: 119283, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38830395

ABSTRACT

BACKGROUND: Animal and human studies indicate that exposure to air pollution and natural environments might modulate the gut microbiota, but epidemiological evidence is very scarce. OBJECTIVES: To assess the potential impact of pre- and postnatal exposure to air pollution and green spaces on infant gut microbiota assembly and trajectories during the first year of life. METHODS: MAMI ("MAternal MIcrobes") birth cohort (Valencia, Spain, N = 162) was used to study the impact of environmental exposure (acute and chronic) on infant gut microbiota during the first year of life (amplicon-based 16S rRNA sequencing). At 7 days and at 1, 6 and 12 months, residential pre- and postnatal exposure to air pollutants (NO2, black carbon -BC-, PM2.5 and O3) and green spaces indicators (NDVI and area of green spaces at 300, 500 and 1000 m buffers) were obtained. For the association between exposures and alpha diversity indicators linear regression models (cross-sectional analyses) and mixed models, including individual as a random effect (longitudinal analyses), were applied. For the differential taxon analysis, the ANCOM-BC package with a log count transformation and multiple-testing corrections were used. RESULTS: Acute exposure in the first week of life and chronic postnatal exposure to NO2 were associated with a reduction in microbial alpha diversity, while the effects of green space exposure were not evident. Acute and chronic (prenatal or postnatal) exposure to NO2 resulted in increased abundance of Haemophilus, Akkermansia, Alistipes, Eggerthella, and Tyzerella populations, while increasing green space exposure associated with increased Negativicoccus, Senegalimassilia and Anaerococcus and decreased Tyzzerella and Lachnoclostridium populations. DISCUSSION: We observed a decrease in the diversity of the gut microbiota and signs of alteration in its composition among infants exposed to higher levels of NO2. Increasing green space exposure was also associated with changes in gut microbial composition. Further research is needed to confirm these findings.


Subject(s)
Air Pollution , Gastrointestinal Microbiome , Gastrointestinal Microbiome/drug effects , Humans , Female , Infant , Spain , Air Pollution/adverse effects , Infant, Newborn , Air Pollutants/analysis , Air Pollutants/toxicity , Birth Cohort , Male , Environmental Exposure/adverse effects , Pregnancy , Cohort Studies , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/microbiology
4.
Proteomics ; 23(23-24): e2300052, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37821362

ABSTRACT

Selenium is a well-known health-relevant element related with cancer chemoprevention, neuroprotective roles, beneficial in diabetes, and in several infectious diseases, among others. It is naturally present in some foods, but deficiency in people led to the production of nutraceuticals, supplements, and functional food enriched in this element. There is a U-shaped link between selenium levels and health and a narrow range between toxic and essential levels, and thus, supplementation should be performed carefully. Omics methodologies have become valuable approaches to delve into the responses of dietary selenium in mammals that allowed a deeper knowledge about the metabolism of this element as well as its biological role. In this review, we discuss omics approaches from the workflows to their applications that has been previously used to deep insight into the metabolism of dietary selenium. There is a special focus on selenoproteins, metabolomics responses in blood and tissues (e.g., brain, reproductive organs, etc.) as well as the impact on gut microbiota and its metabolites profile. Thus, we mainly reviewed heteroatom-tagged proteomics, metallomics, metabolomics, and metataxonomics, usually combined with transcriptomics, genomics, and other molecular methods.


Subject(s)
Gastrointestinal Microbiome , Selenium , Animals , Humans , Selenium/pharmacology , Selenium/metabolism , Dietary Supplements , Proteomics/methods , Genomics , Metabolomics , Mammals/metabolism
5.
Eur J Neurosci ; 58(1): 2215-2231, 2023 07.
Article in English | MEDLINE | ID: mdl-37203224

ABSTRACT

Early life stress (ELS) is associated with metabolic, cognitive, and psychiatric diseases and has a very high prevalence, highlighting the urgent need for a better understanding of the versatile physiological changes and identification of predictive biomarkers. In addition to programming the hypothalamic-pituitary-adrenal (HPA) axis, ELS may also affect the gut microbiota and metabolome, opening up a promising research direction for identifying early biomarkers of ELS-induced (mal)adaptation. Other factors affecting these parameters include maternal metabolic status and diet, with maternal obesity shown to predispose offspring to later metabolic disease. The aim of the present study was to investigate the long-term effects of ELS and maternal obesity on the metabolic and stress phenotype of rodent offspring. To this end, offspring of both sexes were subjected to an adverse early-life experience, and their metabolic and stress phenotypes were examined. In addition, we assessed whether a prenatal maternal and an adult high-fat diet (HFD) stressor further shape observed ELS-induced phenotypes. We show that ELS has long-term effects on male body weight (BW) across the lifespan, whereas females more successfully counteract ELS-induced weight loss, possibly by adapting their microbiota, thereby stabilizing a balanced metabolome. Furthermore, the metabolic effects of a maternal HFD on BW are exclusively triggered by a dietary challenge in adult offspring and are more pronounced in males than in females. Overall, our study suggests that the female microbiota protects against an ELS challenge, rendering them more resilient to additional maternal- and adult nutritional stressors than males.


Subject(s)
Adverse Childhood Experiences , Obesity, Maternal , Prenatal Exposure Delayed Effects , Animals , Mice , Female , Male , Humans , Pregnancy , Obesity/metabolism , Diet, High-Fat/adverse effects , Rodentia , Biomarkers , Prenatal Exposure Delayed Effects/metabolism
6.
Pediatr Res ; 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092964

ABSTRACT

BACKGROUND: Children with cystic fibrosis (CF) present with gut dysbiosis, and current evidence impedes robust recommendations on the use of prebiotics. This study aimed at establishing the prebiotic potential of a commercial beta-glucan on the in vitro colonic microbiota of a child with CF compared to a healthy counterpart (H). METHODS: A dynamic simulator of colonic fermentation (twin-SHIME® model) was set up including the simulation of the proximal (PC) and distal colon (DC) of the CF and the H subjects by colonizing the bioreactors with faecal microbiota. During two weeks the system was supplied with the beta-glucan. At baseline, during treatment and post-treatment, microbiota composition was profiled by 16 S rRNA and short-chain fatty acids (SCFA) production was determined by GS-MS. RESULTS: At baseline, Faecalibacterium, was higher in CF' DC than in the H, along higher Acidaminococcus and less Megasphaera and Sutterella. Beta-glucan supplementation induced increased microbiota richness and diversity in both subjects during the treatment. At genus level, Pseudomonas and Veillonella decreased, while Akkermansia and Faecalibacterium increased significantly in CF. CONCLUSION: The supplementation with beta-glucan suggests positive results on CF colonic microbiota in the in vitro context, encouraging further research in the in vivo setting. IMPACT: Current evidence supports assessing the effect of prebiotics on modifying cystic fibrosis microbiota. The effect of beta-glucan supplementation was evaluated in a controlled dynamic in vitro colonic ecosystem. Beta-glucan supplement improved diversity in cystic fibrosis colonic microbiota. The treatment showed increased abundance of Faecalibacterium and Akkermansia in cystic fibrosis. New evidence supports the use of prebiotics in future clinical studies.

7.
Crit Rev Food Sci Nutr ; 63(26): 7945-7982, 2023.
Article in English | MEDLINE | ID: mdl-35352583

ABSTRACT

Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.


Subject(s)
Breast Feeding , Milk, Human , Infant , Female , Humans , Child , Nutritional Status , Cognition
8.
Int J Mol Sci ; 24(18)2023 Sep 09.
Article in English | MEDLINE | ID: mdl-37762194

ABSTRACT

There is a lack of direct evidence regarding gut microbiota dysbiosis and changes in short-chain fatty acids (SCFAs) in heart failure (HF) patients. We sought to assess any association between gut microbiota composition, SCFA production, clinical parameters, and the inflammatory profile in a cohort of newly diagnosed HF patients. In this longitudinal prospective study, we enrolled eighteen newly diagnosed HF patients. At admission and after 12 months, blood samples were collected for the assessment of proinflammatory cytokines, monocyte populations, and endothelial dysfunction, and stool samples were collected for analysis of gut microbiota composition and quantification of SCFAs. Twelve months after the initial HF episode, patients demonstrated improved clinical parameters and reduced inflammatory state and endothelial dysfunction. This favorable evolution was associated with a reversal of microbiota dysbiosis, consisting of the increment of health-related bacteria, such as genus Bifidobacterium, and levels of SCFAs, mainly butyrate. Furthermore, there was a decrease in the abundance of pathogenic bacteria. In vitro, fecal samples collected after 12 months of follow-up exhibited lower inflammation than samples collected at admission. In conclusion, the favorable progression of HF patients after the initial episode was linked to the reversal of gut microbiota dysbiosis and increased SCFA production, particularly butyrate. Whether restoring butyrate levels or promoting the growth of butyrate-producing bacteria could serve as a complementary treatment for these patients deserves further studies.


Subject(s)
Gastrointestinal Microbiome , Heart Failure , Humans , Dysbiosis , Prospective Studies , Fatty Acids, Volatile , Butyrates
9.
Plant Foods Hum Nutr ; 78(1): 132-138, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36370293

ABSTRACT

Yarrow (Achillea millefolium L., AM) and nettle (Urtica dioica L., UD) are bioactive plants used commercially in functional food and supplement applications and traditionally to alleviate gastric disorders. In this work, the effects of food-grade optimized extracts of Finnish early-season AM and UD were tested on bacterial growth including potential beneficial and foodborne pathogens, as well as murine norovirus (MNV). The anti-inflammatory properties of the extracts were also tested in vitro by NF-κB reporter cells. The food-grade extraction was optimized with the response surface modelling in terms of total carotenoid, chlorophyll, and phenolic compounds contents and antioxidant capacities. The optimal food-grade extraction parameters were a 1-h extraction in 70% ethanol at 45 °C for AM, and at 49 °C for UD. There were no significant effects on the beneficial bacteria (Lacticaseibacillus and Bifidobacterium strains), and the extracts were more effective against gram-positive than gram-negative foodborne bacteria and potential pathogens. Listeria innocua was the most susceptible strain in the optimized extracts with a growth rate of 0.059 ± 0.004 for AM and 0.067 ± 0.006 for UD, p < 0.05 compared to control. The optimized extracts showed a logarithmic growth reduction of 0.67 compared to MNV. The hydroethanolic extracts were cytotoxic to both cell lines, whereas aqueous AM and UD extracts induced and reduced TLR4 signalling in a reporter cell line, respectively. The results provide novel food-grade extraction parameters and support the bioactive effects of AM and UD in functional food applications, but more research is needed to elucidate the precise biological activity in vivo for gastric health.


Subject(s)
Achillea , Urtica dioica , Mice , Animals , Plant Extracts/pharmacology , Plant Leaves , Antioxidants/pharmacology , Bacteria
10.
J Proteome Res ; 21(3): 758-767, 2022 03 04.
Article in English | MEDLINE | ID: mdl-34734730

ABSTRACT

Selenium (Se) is an essential trace element with important health roles due to the antioxidant properties of selenoproteins. To analyze the interplay between Se and gut microbiota, gut metabolomic profiles were determined in conventional (C) and microbiota depleted mice (Abx) after Se-supplementation (Abx-Se) by untargeted metabolomics, using an analytical multiplatform based on GC-MS and UHPLC-QTOF-MS (MassIVE ID MSV000087829). Gut microbiota profiling was performed by 16S rRNA gene amplicon sequencing. Significant differences in the levels of about 70% of the gut metabolites determined, including fatty acyls, glycerolipids, glycerophospholipids, and steroids, were found in Abx-Se compared to Abx, and only 30% were different between Abx-Se and C, suggesting an important effect of Se-supplementation on Abx mice metabolism. At genus level, the correlation analysis showed strong associations between metabolites and gut bacterial profiles. Likewise, higher abundance of Lactobacillus spp., a potentially beneficial genus enriched after Se-supplementation, was associated with higher levels of prenol lipids, phosphatidylglycerols (C-Se), steroids and diterpenoids (Abx-Se), and also with lower levels of fatty acids (Abx-Se). Thus, we observed a crucial interaction between Se intake-microbiota-metabolites, although further studies to clarify the specific mechanisms are needed. This is the first study about untargeted gut metabolomics after microbiota depletion and Se-supplementation.


Subject(s)
Gastrointestinal Microbiome , Selenium , Animals , Dietary Supplements , Gastrointestinal Microbiome/genetics , Metabolomics , Mice , RNA, Ribosomal, 16S/genetics , Selenium/pharmacology
11.
FASEB J ; 35(7): e21649, 2021 07.
Article in English | MEDLINE | ID: mdl-34164846

ABSTRACT

Eating chocolate in the morning or in the evening/at night, may differentially affect energy balance and impact body weight due to changes in energy intake, substrate oxidation, microbiota (composition/function), and circadian-related variables. In a randomized controlled trial, postmenopausal females (n = 19) had 100 g of chocolate in the morning (MC), in the evening/at night (EC), or no chocolate (N) for 2 weeks and ate any other food ad libitum. Our results show that 14 days of chocolate intake did not increase body weight. Chocolate consumption decreased hunger and desire for sweets (P < .005), and reduced ad libitum energy intake by ~300 kcal/day during MC and ~150 kcal/day during EC (P = .01), but did not fully compensate for the extra energy contribution of chocolate (542 kcal/day). EC increased physical activity by +6.9%, heat dissipation after meals +1.3%, and carbohydrate oxidation by +35.3% (P < .05). MC reduced fasting glucose (4.4%) and waist circumference (-1.7%) and increased lipid oxidation (+25.6%). Principal component analyses showed that both timings of chocolate intake resulted in differential microbiota profiles and function (P < .05). Heat map of wrist temperature and sleep records showed that EC induced more regular timing of sleep episodes with lower variability of sleep onset among days than MC (60 min vs 78 min; P = .028). In conclusion, having chocolate in the morning or in the evening/night results in differential effects on hunger and appetite, substrate oxidation, fasting glucose, microbiota (composition and function), and sleep and temperature rhythms. Results highlight that the "when" we eat is a relevant factor to consider in energy balance and metabolism.


Subject(s)
Appetite/drug effects , Body Mass Index , Carbohydrates/chemistry , Chocolate/adverse effects , Hunger/drug effects , Microbiota/drug effects , Aged , Blood Glucose/analysis , Cross-Over Studies , Energy Intake , Fasting , Female , Humans , Middle Aged , Postprandial Period , Time Factors
12.
Brain Behav Immun ; 100: 311-320, 2022 02.
Article in English | MEDLINE | ID: mdl-34920092

ABSTRACT

Maternal gestational obesity is a risk factor for offspring's neurodevelopment and later neuro-cognitive disorders. Altered gut microbiota composition has been found in patients with neurocognitive disorders, and in relation to maternal metabolic health. We explored the associations between gut microbiota and cognitive development during infancy, and their link with maternal obesity. In groups of children from the Pisa birth Cohort (PISAC), we analysed faecal microbiota composition by 16S rRNA marker gene sequencing of first-pass meconium samples and of faecal samples collected at age 3, 6, 12, 24, 36 months, and its relationship with maternal gestational obesity or diabetes, and with cognitive development, as measured from 6 to 60 months of age by the Griffith's Mental Development Scales. Gut microbiota composition in the first phases of life is dominated by Bifidobacteria (Actinobacteria phylum), with contribution of Escherichia/Shigella and Klebsiella genera (Proteobacteria phylum), whereas Firmicutes become more dominant at 36 months of age. Maternal overweight leads to lower abundance of Bifidobacterium, Blautia and Ruminococcus, and lower practical reasoning scores in the offspring at the age of 36 months. In the whole population, microbiota in the first-pass meconium samples shows much higher alpha diversity compared to later samples, and its composition, particularly Bifidobacterium and Veillonella abundances, correlates with practical reasoning scores at 60 months of age. Maternal overweight correlates with bacterial colonization and with the development of reasoning skills at pre-school age. Associations between neonatal gut colonization and later cognitive function provide new perspectives of primary (antenatal) prevention of neurodevelopmental disorders.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Child , Child, Preschool , Cognition , Female , Gastrointestinal Microbiome/genetics , Humans , Infant, Newborn , Overweight , Pregnancy , RNA, Ribosomal, 16S/genetics
13.
Pediatr Res ; 91(7): 1804-1811, 2022 06.
Article in English | MEDLINE | ID: mdl-34349229

ABSTRACT

BACKGROUND: Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS: Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS: Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS: The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT: The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.


Subject(s)
Gastrointestinal Microbiome , Premature Birth , Female , Humans , Iatrogenic Disease , Infant , Infant, Newborn , Infant, Premature , Pregnancy , RNA, Ribosomal, 16S/genetics
14.
J Nutr ; 151(2): 330-340, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33188413

ABSTRACT

BACKGROUND: Breast milk is a complex biofluid that provides nutrients and bioactive agents, including bacteria, for the development of the infant gut microbiota. However, the impact of maternal diet and other factors, such as mode of delivery and antibiotic exposure, on the breast milk microbiota has yet to be understood. OBJECTIVES: This study aimed to examine the association between maternal diet and breast milk microbiota and to ascertain the potential role of mode of delivery and antibiotic exposure. METHODS: In a cross-sectional study of the MAMI cohort, breast milk microbiota profiling was assessed in 120 samples from healthy mothers by 16S rRNA gene sequencing. Maternal dietary information was recorded through an FFQ, and clinical characteristics, including mode of delivery, antibiotic exposure, and exclusive breastfeeding, were collected. RESULTS: Maternal diet was grouped into 2 clusters: Cluster I (high intake of plant protein, fiber, and carbohydrates), and Cluster II (high intake of animal protein and lipids). Breast milk microbiota was shaped by maternal dietary clusters. Staphylococcus and Bifidobacterium were associated with carbohydrate intake whereas the Streptococcus genus was associated with intakes of the n-3 PUFAs [EPA and docosapentaenoic acid (22:5ω-3)]. Mode of delivery and antibiotic exposure influenced breast milk microbiota in a diet cluster-dependent manner. Differences between/among the maternal dietary clusters were found in the milk microbiota of the cesarean-section (C-section)/antibiotic group, whereas no differences were observed in vaginal births. Lower abundances of Lactobacillus, Bacteroides, and Sediminibacterium genera were observed in Cluster II/C-section/antibiotic exposure compared with the other groups. CONCLUSIONS: Maternal diet shapes the composition and diversity of breast milk microbiota, with the most important contributions coming from dietary fiber and both plant and animal protein intakes. The relation between the maternal diet and the milk microbiota needs further research because it has a key impact on infant microbiota development and contributes to infant health outcomes in the short and long term.This trial was registered at clinicaltrials.gov as NCT03552939.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Bacteria/classification , Diet , Microbiota/drug effects , Milk, Human/chemistry , Milk, Human/microbiology , Adult , Bacteria/drug effects , Cohort Studies , Cross-Sectional Studies , Female , Humans , Infant , Infant, Newborn , Male
15.
BMC Infect Dis ; 21(1): 373, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33882835

ABSTRACT

BACKGROUND: Aberrant microbiota composition has been linked to disease development at numerous anatomical sites. Microbiota changes in reaction to viral infections, such as human papillomavirus (HPV), have been investigated almost exclusively in the female reproductive tract. However, HPV infection may also affect male health by reducing semen quality and fertility. The aim of this study was to investigate whether present HPV DNA is associated with detectable changes in semen bacterial microbiota composition and diversity. METHODS: This study relied on stored semen samples from 31 fertile healthy men who participated in the Finnish family HPV Study during the years 1998-2001. DNA was extracted from semen with PCR template preparation kit. HPV was genotyped using Luminex-based Multimetrix® assay. Microbiota was analyzed from the V3-V4 region of 16S rDNA gene following sequencing on an Illumina MiSeq platform. All statistical analyses were performed with Calypso software version 8.84. RESULTS: HPV DNA was detected in 19.4% (6/31) of the semen samples. HPV status in the semen did not impact the α-diversity estimations, as measured by Chao1 and Shannon indices, nor ß-diversity. Nevertheless, HPV-positive semen samples exhibited differences in the taxonomic composition of the bacterial microbiota including higher abundances of Moraxellaceae (p = 0.028), Streptococcus (p = 0.0058) and Peptostreptococcus (p = 0.012) compared to HPV-negative semen samples. CONCLUSION: HPV infection is associated with altered bacterial microbiota composition in semen, and this might have in impact to male health in general. As of present, it is unclear whether these changes result from HPV infection or whether altered bacterial microbiota increases susceptibility to HPV infection. More research is needed on viral-bacterial interactions in the male reproductive system.


Subject(s)
Bacteria/genetics , Microbiota/genetics , Papillomaviridae/genetics , Papillomavirus Infections/epidemiology , Semen/microbiology , Adult , DNA, Ribosomal/genetics , DNA, Viral/genetics , Female , Finland/epidemiology , Genotype , Healthy Volunteers , High-Throughput Nucleotide Sequencing , Humans , Male , Papillomavirus Infections/virology , Polymerase Chain Reaction , Semen Analysis , Young Adult
16.
Mar Drugs ; 19(4)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33916965

ABSTRACT

Fishery by-products are rich in biologically active substances and the use of green and efficient extraction methods to recover these high-added-value compounds is of particular importance. In this study, head, skin and viscera of rainbow trout and sole were used as the target matrices and accelerated solvent extraction (ASE) (45-55 °C, 15 min, pH 5.2-6.8, 103.4 bars) and pulsed electric fields (PEF) (1-3 kV/cm, 123-300 kJ/kg, 15-24 h) were applied as extraction technologies. The results showed that ASE and PEF significantly increased the protein extract efficiency of the fish by-products (p < 0.05) by up to 80%. SDS-PAGE results showed that ASE and PEF treatments changed the molecular size distribution of the protein in the extracts, which was specifically expressed as the change in the area or number of bands between 5 and 250 kDa. The antioxidant capacity of the extracts was evaluated by oxygen radical absorbance capacity (ORAC) and total antioxidant capacity (ABTS) assays. The results showed that both ASE and PEF treatments significantly increased the antioxidant capacity of rainbow trout and sole skin and head extracts (p < 0.05). ASE and PEF extraction processes can be used as new technologies to extract high-added-value compounds from fish by-products.


Subject(s)
Antioxidants/pharmacology , Electricity , Fish Proteins/pharmacology , Flatfishes/metabolism , Oncorhynchus mykiss/metabolism , Seafood , Solvents/chemistry , Animals , Antioxidants/isolation & purification , Chemical Fractionation , Electrophoresis, Polyacrylamide Gel , Fish Proteins/isolation & purification , Food Handling , Hydrogen-Ion Concentration , Molecular Weight , Oxygen Radical Absorbance Capacity , Pressure , Temperature , Time Factors , Waste Products
17.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498321

ABSTRACT

The gut microbiota has emerged as a key factor in the pathogenesis of intestinal viruses, including enteroviruses, noroviruses and rotaviruses (RVs), where stimulatory and inhibitory effects on infectivity have been reported. With the aim of determining whether members of the microbiota interact with RVs during infection, a combination of anti-RV antibody labeling, fluorescence-activated cell sorting and 16S rRNA amplicon sequencing was used to characterize the interaction between specific bacteria and RV in stool samples of children suffering from diarrhea produced by G1P[8] RV. The genera Ruminococcus and Oxalobacter were identified as RV binders in stools, displaying enrichments between 4.8- and 5.4-fold compared to samples nonlabeled with anti-RV antibodies. In vitro binding of the G1P[8] Wa human RV strain to two Ruminococcus gauvreauii human isolates was confirmed by fluorescence microscopy. Analysis in R. gauvreauii with antibodies directed to several histo-blood group antigens (HBGAs) indicated that these bacteria express HBGA-like substances on their surfaces, which can be the target for RV binding. Furthermore, in vitro infection of the Wa strain in differentiated Caco-2 cells was significantly reduced by incubation with R. gauvreauii. These data, together with previous findings showing a negative correlation between Ruminococcus levels and antibody titers to RV in healthy individuals, suggest a pivotal interaction between this bacterial group and human RV. These results reveal likely mechanisms of how specific bacterial taxa of the intestinal microbiota could negatively affect RV infection and open new possibilities for antiviral strategies.


Subject(s)
Gastrointestinal Microbiome , Rotavirus Infections/microbiology , Rotavirus/metabolism , Ruminococcus/metabolism , Bacterial Proteins/metabolism , Caco-2 Cells , Child, Preschool , Humans , Intestines/microbiology , Intestines/virology , Protein Binding , Rotavirus/pathogenicity , Rotavirus Infections/virology , Ruminococcus/pathogenicity
18.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33579027

ABSTRACT

Mothers confer natural passive immunization to their infants through the transplacental pathway during the gestation period. The objective of the present study was to establish at birth the maternal and cord plasma concentration and relationship of immunoglobulins (Igs), cytokines (CKs), and adipokines. In addition, the impact of the maternal microbiota and diet was explored. The plasma profile of these components was different between mothers and babies, with the levels of many CKs, IgM, IgG2a, IgE, IgA, and leptin significantly higher in mothers than in the cord sample. Moreover, the total Igs, all IgG subtypes, IgE, and the Th1/Th2 ratio positively correlated in the mother-infant pair. Maternal dietary components such as monounsaturated fatty acids-polyunsaturated fatty acids and fiber were positively associated with some immune factors such as IgA in cord samples. The microbiota composition clustering also influenced the plasma profile of some factors (i.e., many CKs, some Ig, and adiponectin). In conclusion, we have established the concentration of these immunomodulatory factors in the maternal-neonatal pair at birth, some positive associations, and the influence of maternal diet and the microbiota composition, suggesting that the immune status during pregnancy, in terms of CKs and Igs levels, can influence the immune status of the infant at birth.


Subject(s)
Cytokines/blood , Diet , Fetal Blood , Immunoglobulins/blood , Microbiota , Adipokines/blood , Adipokines/immunology , Cytokines/immunology , Feces/microbiology , Female , Fetal Blood/chemistry , Fetal Blood/immunology , Humans , Immunity , Immunoglobulins/immunology , Infant, Newborn , Male , Nutritional Status , Pregnancy
19.
Pediatr Res ; 88(1): 57-65, 2020 07.
Article in English | MEDLINE | ID: mdl-31261372

ABSTRACT

BACKGROUND: There is currently a lack of experimental evidence for horizontal gene transfer (HGT) mechanisms in the human gut microbiota. The aim of this study was therefore to experimentally determine the HGT potential in the microbiota of a healthy preterm infant twin pair and to evaluate the global occurrence of the mobilized elements. METHODS: Stool samples were collected. Both shotgun metagenome sequencing and bacterial culturing were done for the same samples. A range of experimental conditions were used to test DNA transfer for the cultured isolates. Searches for global distribution of transferable elements were done for the ~120,000 metagenomic samples in the Sequence Read Archive (SRA) database. RESULTS: DNA transfer experiments demonstrated frequent transmission of an ESBL encoding IncI1 plasmid, a high copy number ColEI plasmid, and bacteriophage P1. Both IncI1 and ColE1 were abundant in the stool samples. In vitro competition experiments showed that transconjugants containing IncI1 plasmids outcompeted the recipient strain in the absence of antibiotic selection. The SRA searches indicated a global distribution of the mobilizable elements, with chicken identified as a possible reservoir for the IncI1 ESBL encoding plasmid. CONCLUSION: Our results experimentally support a major horizontal transmission and persistence potential of the preterm infant gut microbiota mobilome involving genes encoding ESBL.


Subject(s)
Gastrointestinal Microbiome , Gene Transfer Techniques , Gene Transfer, Horizontal , Multigene Family , Animals , Anti-Bacterial Agents , Bacteriophages , Chickens , Contig Mapping , DNA Transposable Elements , DNA, Bacterial/analysis , Enterococcus/genetics , Escherichia coli/genetics , Humans , Infant, Newborn , Infant, Premature , Plasmids/genetics , Prevalence , Prospective Studies , Sequence Analysis, DNA , Staphylococcus epidermidis/genetics , Twins
20.
Pediatr Allergy Immunol ; 31(3): 250-257, 2020 04.
Article in English | MEDLINE | ID: mdl-31736150

ABSTRACT

BACKGROUND: Early colonization with a diverse microbiota seems to play a crucial role for appropriate immune maturation during childhood. Breastmilk microbiota is one important source of microbes for the infant, transferred together with maternal IgA antibodies. We previously observed that allergy development during childhood was associated with aberrant IgA responses to the gut microbiota already at 1 month of age, when the IgA antibodies are predominantly maternally derived in breastfed infants. OBJECTIVE: To determine the microbial composition and IgA-coated bacteria in breastmilk in relation to allergy development in children participating in an intervention trial with pre- and post-natal Lactobacillus reuteri supplementation. METHODS: A combination of flow cytometric cell sorting and 16S rRNA gene sequencing was used to characterize the bacterial recognition patterns by IgA in breastmilk samples collected one month post-partum from 40 mothers whose children did or did not develop allergic and asthmatic symptoms during the first 7 years of age. RESULTS: The milk fed to children developing allergic manifestations had significantly lower bacterial richness, when compared to the milk given to children that remained healthy. Probiotic treatment influenced the breastmilk microbiota composition. However, the proportions of IgA-coated bacteria, the total bacterial load and the patterns of IgA-coating were similar in breastmilk between mothers of healthy children and those developing allergies. CONCLUSION: Consumption of breastmilk with a reduced microbial richness in the first month of life may play an important role in allergy development during childhood.


Subject(s)
Hypersensitivity/epidemiology , Milk, Human/immunology , Milk, Human/microbiology , Asthma/epidemiology , Breast Feeding , Child , Child, Preschool , Female , Flow Cytometry , Food Hypersensitivity/epidemiology , Gastrointestinal Microbiome , Humans , Hypersensitivity/immunology , Hypersensitivity/microbiology , Immunoglobulin A/immunology , Infant , Infant, Newborn , Male , Microbiota/genetics , Probiotics/therapeutic use , RNA, Ribosomal, 16S
SELECTION OF CITATIONS
SEARCH DETAIL