Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.318
Filter
1.
Front Cell Infect Microbiol ; 14: 1428525, 2024.
Article in English | MEDLINE | ID: mdl-39310784

ABSTRACT

Introduction: Managing burn injuries is a challenge in healthcare. Due to the alarming increase in antibiotic resistance, new prophylactic and therapeutic strategies are being sought. This study aimed to evaluate the potential of live Lactic Acid Bacteria for managing burn infections, using Galleria mellonella larvae as an alternative preclinical animal model and comparing the outcomes with a common antibiotic. Methods: The antimicrobial activity of LAB isolated from human breast milk was assessed in vitro against Pseudomonas aeruginosa ATCC 27853. Additionally, the immunomodulatory effects of LAB were evaluated in vivo using the G. mellonella burn wound infection model. Results and discussion: In vitro results demonstrated the antimicrobial activity of Lactic Acid Bacteria against P. aeruginosa. In vivo results show that their prophylactic treatment improves, statistically significant, larval survival and modulates the expression of immunity-related genes, Gallerimycin and Relish/NF-κB, strain-dependently. These findings lay the foundation and suggest a promising alternative for burn wound prevention and management, reducing the risk of antibiotic resistance, enhancing immune modulation, and validating the potential G. mellonella as a skin burn wound model.


Subject(s)
Burns , Disease Models, Animal , Lactobacillales , Larva , Milk, Human , Pseudomonas aeruginosa , Animals , Burns/microbiology , Pseudomonas aeruginosa/drug effects , Humans , Larva/microbiology , Milk, Human/microbiology , Female , Pseudomonas Infections/microbiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/immunology , Moths/microbiology , Wound Infection/microbiology , Wound Infection/drug therapy , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests
2.
Microbiome ; 12(1): 182, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342403

ABSTRACT

BACKGROUND: Children born to women with HIV but who do not become HIV infected experience increased morbidity and mortality compared with children born to women without HIV. The basis of this increased vulnerability is unknown. The microbiome, specifically the infant gut microbiome, likely plays an important role in infant immune development. The human milk microbiome is thought to have an important role in the development of the infant gut and therefore, if perturbed, may contribute to this increased vulnerability. We investigated the effects of HIV and its therapies on the milk microbiome and possible changes in the milk microbiome before or after infant HIV infection. RESULTS: Seven-hundred fifty-six human milk samples were selected from three separate studies conducted over a 15-year period to investigate the role of HIV and its therapies on the human milk microbiome. Our data reveal that the milk microbiome is modulated by parity (R2 = 0.006, p = 0.041), region/country (R2 = 0.014, p = 0.007), and duration of lactation (R2 = 0.027-0.038, all p < 0.001). There is no evidence, however, using 16S rRNA V4 amplicon sequencing, that the human milk microbiome is altered by HIV infection (R2 = 0.003, p = 0.896), by combination antiretroviral therapy (R2 = 0.0009, p = 0.909), by advanced maternal disease (R2 = 0.003, p = 0.263), or in cases of infant infection either through isolated early mucosal (R2 = 0.003, p = 0.197) or early mucosal and breast milk transmission (R2 = 0.002, p = 0.587). CONCLUSIONS: The milk microbiome varies by stage of lactation, by parity, and by region; however, we found no evidence that the human milk microbiome is altered by maternal HIV infection, disease severity, or antiretroviral therapy. Additionally, we found no association between the milk microbiome and transmission of HIV to the infant. Investigations including higher resolution microbiome approaches or into other potential mechanisms to understand why the approximately one million children born annually to women with HIV escape infection, but do not escape harm, are urgently needed. Video Abstract.


Subject(s)
HIV Infections , Milk, Human , RNA, Ribosomal, 16S , Humans , Milk, Human/microbiology , Milk, Human/virology , HIV Infections/microbiology , HIV Infections/virology , Female , RNA, Ribosomal, 16S/genetics , Pregnancy , Infectious Disease Transmission, Vertical , Infant , Microbiota , Gastrointestinal Microbiome , Infant, Newborn , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/virology , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Adult , Breast Feeding , Lactation
3.
BMC Microbiol ; 24(1): 350, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289612

ABSTRACT

Diarrheal diseases remain the leading cause of high mortality among the infants, particularly in the developing countries; Probiotic intervention for diarrhea has been an ongoing novel approach to diarrheal prevention and treatment. This study aims to characterize immunogenic and probiotic properties of lactic acid bacteria (LAB) isolated from human breast milk and neonates' faeces. The LAB isolates from 16 mothers' breast milk and 13 infants' faeces were screened and identified by 16 S rRNA gene partial sequencing. Their antimicrobial activities against 5 strains of diarrheagenic Escherichia coli were tested. Organic acids production was quantified by HPLC, and antibiotic resistance pattern were determined by VITEK®. Autoaggregation, co-aggregation and hydrophobicity properties were assessed by UV spectrophotometry and immunomodulatory effect was determined in mouse model. Ninety-three LAB of five genera were identified. The most abundant species was Lactiplantibacillus plantarum with inhibition zones ranged from 8.0 to 25.0 ± 1 mm. Lacticaseibacillus rhamnosus A012 had 76.8 mg/mL lactic acid, (the highest concentration), was susceptible to all antibiotics tested. L. plantarum A011 and L. rhamnosus A012 were highly resistance to gastrointestinal conditions. L. rhamnosus A012 produced hydrophobicity of 25.01% (n-hexadecane), 15.4% (xylene) and its autoaggregation was 32.52%. L. rhamnosus A012 and L. plantarum A011 exert immunomodulatory effects on the cyclophosphamide-treated mice by upregulating anti-inflammatory cytokine and downregulating proinflammatory cytokines. Lactobacillus sp. demonstrated good probiotic and immunomodulatory properties. Further works are ongoing on the practical use of the strains.


Subject(s)
Diarrhea , Escherichia coli , Feces , Lactobacillales , Milk, Human , Probiotics , Probiotics/pharmacology , Humans , Feces/microbiology , Animals , Female , Milk, Human/microbiology , Milk, Human/immunology , Mice , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/immunology , Lactobacillales/isolation & purification , Lactobacillales/physiology , Lactobacillales/classification , Diarrhea/microbiology , Diarrhea/prevention & control , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Infant , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/pharmacology , Infant, Newborn , Adult , Microbial Sensitivity Tests
4.
Cell ; 187(19): 5431-5452.e20, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39303691

ABSTRACT

Breastfeeding and microbial colonization during infancy occur within a critical time window for development, and both are thought to influence the risk of respiratory illness. However, the mechanisms underlying the protective effects of breastfeeding and the regulation of microbial colonization are poorly understood. Here, we profiled the nasal and gut microbiomes, breastfeeding characteristics, and maternal milk composition of 2,227 children from the CHILD Cohort Study. We identified robust colonization patterns that, together with milk components, predict preschool asthma and mediate the protective effects of breastfeeding. We found that early cessation of breastfeeding (before 3 months) leads to the premature acquisition of microbial species and functions, including Ruminococcus gnavus and tryptophan biosynthesis, which were previously linked to immune modulation and asthma. Conversely, longer exclusive breastfeeding supports a paced microbial development, protecting against asthma. These findings underscore the importance of extended breastfeeding for respiratory health and highlight potential microbial targets for intervention.


Subject(s)
Breast Feeding , Milk, Human , Humans , Female , Milk, Human/microbiology , Infant , Child, Preschool , Asthma/microbiology , Asthma/prevention & control , Asthma/immunology , Microbiota , Gastrointestinal Microbiome , Male , Cohort Studies , Infant, Newborn
5.
BMC Res Notes ; 17(1): 271, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289764

ABSTRACT

OBJECTIVE: In this pilot study, we verified safe practices for breast milk expression, storage, and duration, based on bacteriological results. RESULTS: We collected breast milk samples from three healthy lactating volunteers and analyzed the bacterial flora and changes in the viable bacterial counts (including those of Staphylococcus) of the samples. Although no consistent change could be observed in the abundance of a particular bacterial group in samples expressed under hygienic control conditions, viable bacterial counts were higher in self-expressed milk than in milk expressed under hygienic control conditions. In conclusion, increased hygiene awareness is vital during breast milk expression and storage.


Subject(s)
Milk, Human , Humans , Milk, Human/microbiology , Pilot Projects , Female , Hygiene/standards , Adult , Breast Milk Expression , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , Staphylococcus/isolation & purification , Lactation
6.
NPJ Biofilms Microbiomes ; 10(1): 85, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39277573

ABSTRACT

The gut microbiota of infants in low- to middle-income countries is underrepresented in microbiome research. This study explored the faecal microbiota composition and faecal cytokine profiles in a cohort of infants in a rural province of Cambodia and investigated the impact of sample storage conditions and infant environment on microbiota composition. Faecal samples collected at three time points from 32 infants were analysed for microbiota composition using 16S rRNA amplicon sequencing and concentrations of faecal cytokines. Faecal bacterial isolates were subjected to whole genome sequencing and genomic analysis. We compared the effects of two sample collection methods due to the challenges of faecal sample collection in a rural location. Storage of faecal samples in a DNA preservation solution preserved Bacteroides abundance. Microbiota analysis of preserved samples showed that Bifidobacterium was the most abundant genus with Bifidobacterium longum the most abundant species, with higher abundance in breast-fed infants. Most infants had detectable pathogenic taxa, with Shigella and Klebsiella more abundant in infants with recent diarrhoeal illness. Neither antibiotics nor infant growth were associated with gut microbiota composition. Genomic analysis of isolates showed gene clusters encoding the ability to digest human milk oligosaccharides in B. longum and B. breve isolates. Antibiotic-resistant genes were present in both potentially pathogenic species and in Bifidobacterium. Faecal concentrations of Interlukin-1alpha and vascular endothelial growth factor were higher in breast-fed infants. This study provides insights into an underrepresented population of rural Cambodian infants, showing pathogen exposure and breastfeeding impact gut microbiota composition and faecal immune profiles.


Subject(s)
Bifidobacterium , Cytokines , Diarrhea , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Rural Population , Humans , Feces/microbiology , Infant , Cambodia , Cytokines/metabolism , RNA, Ribosomal, 16S/genetics , Female , Male , Diarrhea/microbiology , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Diet , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Shigella/genetics , Shigella/isolation & purification , Bacteroides/genetics , Bacteroides/isolation & purification , Klebsiella/genetics , Klebsiella/isolation & purification , Breast Feeding , DNA, Bacterial/genetics , Whole Genome Sequencing , Milk, Human/microbiology , Milk, Human/chemistry
7.
Cell Rep Med ; 5(9): 101729, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39243753

ABSTRACT

Mother's milk contains diverse bacterial communities, although their impact on microbial colonization in very-low-birth-weight (VLBW, <1,500 g) infants remains unknown. Here, we examine relationships between the microbiota in preterm mother's milk and the VLBW infant gut across initial hospitalization (n = 94 mother-infant dyads, 422 milk-stool pairs). Shared zero-radius operational taxonomic units (zOTUs) between milk-stool pairs account for ∼30%-40% of zOTUs in the VLBW infant's gut. We show dose-response relationships between intakes of several genera from milk and their concentrations in the infant's gut. These relationships and those related to microbial sharing change temporally and are modified by in-hospital feeding practices (especially direct breastfeeding) and maternal-infant antibiotic use. Correlations also exist between milk and stool microbial consortia, suggesting that multiple milk microbes may influence overall gut communities together. These results highlight that the mother's milk microbiota may shape the gut colonization of VLBW infants by delivering specific bacteria and through intricate microbial interactions.


Subject(s)
Feces , Gastrointestinal Microbiome , Infant, Very Low Birth Weight , Milk, Human , Milk, Human/microbiology , Humans , Gastrointestinal Microbiome/physiology , Female , Infant, Newborn , Feces/microbiology , Microbial Consortia , Breast Feeding , Adult , Male , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Infant, Premature , Mothers
8.
Bratisl Lek Listy ; 125(10): 605-611, 2024.
Article in English | MEDLINE | ID: mdl-39344763

ABSTRACT

BACKGROUND: Human milk oligosaccharide (HMO) is a unique component of breastmilk. To date, no study has investigated the correlation between HMO and infant nutritional status particularly through the lens of gut microbiota. Therefore, our study aims to investigate the relationships between 2'-Fucosyllactose (2'-FL) in HMO and Firmicutes/Bacteroidetes (F/B) ratio among stunted infants. METHODS: A case-control study was conducted among 103 mother-infant pairs in Malang City, Indonesia. The quantification of 2'-FL HMO was assessed using High-Performance Liquid Chromatography (HPLC). The F/B ratio was analyzed with real-time poly-chain reaction (RT-PCR). For bivariate analysis, we employed the Spearman correlation and Mann‒Whitney tests, while for multivariate analysis, we utilized multiple linear regression. RESULTS: The findings showed that the stunted nutritional status was detected in 49 out of 103 infants. In this group, 40.81% of mothers of infants with a stunted nutritional status had a secretor-positive status, while all mothers of infants with appropriate nutritional status tested positive for the secretor status (100%). However, the association between maternal secretor status and infant nutritional status was not statistically significant (p>0.05). The average levels of 2'-FL HMO in breast milk were lower in the group with stunted infants compared to non-stunted infants (1.21 mg/L vs 1.40 mg/L). The regression analysis revealed a significant association of 2'-FL HMO levels with the presence of Bacteroidetes and value of the F/B ratio (p>0.05). CONCLUSIONS: The breast milk component 2'-FL HMO significantly influences the gut microbiota of stunted infants. Future research aimed at elucidating the mechanisms by which 2'-FL HMO modulates infant gut microbiota should consider not only concentration and specific bacterial taxa but also intake levels (Tab. 2, Fig. 1, Ref. 37). Text in PDF www.elis.sk Keywords: 2'-fucosyllactose, human milk, oligosaccharide, firmicutes, bacteroidetes, stunting, infant.


Subject(s)
Bacteroidetes , Firmicutes , Milk, Human , Oligosaccharides , Humans , Milk, Human/chemistry , Milk, Human/microbiology , Indonesia , Female , Oligosaccharides/analysis , Oligosaccharides/metabolism , Case-Control Studies , Infant , Bacteroidetes/isolation & purification , Firmicutes/isolation & purification , Trisaccharides/analysis , Male , Adult , Nutritional Status , Gastrointestinal Microbiome , Infant, Newborn
9.
Compr Rev Food Sci Food Saf ; 23(5): e13431, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39165140

ABSTRACT

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


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Oligosaccharides , Prebiotics , Humans , Gastrointestinal Microbiome/physiology , Milk, Human/chemistry , Milk, Human/microbiology , Oligosaccharides/chemistry , Probiotics , Infant , Bacteria/metabolism , Bifidobacterium/physiology
10.
Gut Microbes ; 16(1): 2392009, 2024.
Article in English | MEDLINE | ID: mdl-39161102

ABSTRACT

Here, we explored the vast potential of microbiome-based interventions in preventing and managing non-communicable diseases including obesity, diabetes, allergies, celiac disease, inflammatory bowel diseases, malnutrition, and cardiovascular diseases across different life stages. We discuss the intricate relationship between microbiome and non-communicable diseases, emphasizing on the "window of opportunity" for microbe-host interactions during the first years after birth. Specific biotics and also live biotherapeutics including fecal microbiota transplantation emerge as pivotal tools for precision medicine, acknowledging the "one size doesn't' fit all" aspect. Challenges in implementation underscore the need for advanced technologies, scientific transparency, and public engagement. Future perspectives advocate for understanding maternal-neonatal microbiome, exploring the maternal exposome and delving into human milk's role in the establishment and restoration of the infant microbiome and its influence over health and disease. An integrated scientific approach, employing multi-omics and accounting for inter-individual variance in microbiome composition and function appears central to unleash the full potential of early-life microbiome interventions in revolutionizing healthcare.


Subject(s)
Gastrointestinal Microbiome , Humans , Pregnancy , Female , Infant, Newborn , Milk, Human/microbiology , Fecal Microbiota Transplantation , Infant , Host Microbial Interactions
11.
Sci Rep ; 14(1): 19199, 2024 08 19.
Article in English | MEDLINE | ID: mdl-39160300

ABSTRACT

The main purpose was to determine the abundance of dominant phyla, Bifidobacterium spp., and Lactobacillus in breast milk of obese mothers versus normal-weights in fourth month of lactation in Iranian population. Sixty health women at the fourth month of breastfeeding, aged 18-40 years, were included and categorized based on body mass index (BMI) to the obese (BMI ≥ 30 kg/m2) and normal-weights (18.5 ≤ BMI ≤ 24.9). Bacterial DNA was extracted and qPCR of the 16S region was performed after human milk donation in a sterile condition. A multiple linear mixed model was used to determine the effective factors on the phyla population. Bifidobacterium spp. was significantly higher in milk of normal-weight group than the obese. The current weight showed a significant effect on the Actinobacteria abundance in milk. The Bacteroidetes and Firmicutes were significantly lower in mother's milk with cesarean section (p = 0.04). Pre-pregnancy obesity decreased the Firmicutes and Lactobacillus abundance in maternal milk (p = 0.04 and p = 0.01). The Actinobacteria and Bifidobacterium spp. showed a significant effect on infant's height (p = 0.008 and p = 0.04). The maternal current and pre-pregnancy weight showed an important effect on abundance of Actinobacteria and Bifidobacterium spp., as the good phyla and genus in milk which are associated with the infant's height.


Subject(s)
Lactation , Milk, Human , Obesity , Probiotics , Humans , Female , Milk, Human/microbiology , Adult , Obesity/microbiology , Young Adult , Adolescent , Bifidobacterium/isolation & purification , Bifidobacterium/genetics , Breast Feeding , Body Mass Index , Lactobacillus/isolation & purification , Lactobacillus/genetics , Pregnancy , Iran
12.
Medicina (Kaunas) ; 60(8)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39202589

ABSTRACT

Background and objectives: The development of the oral microbiome begins in the prenatal stage. Breast milk contains antimicrobial proteins, microorganisms, metabolites, enzymes, and immunoglobulins, among others; therefore, differences have been noted in the type of microorganisms that colonize the oral cavity of children who are breastfed compared to those who are formula-fed. Our objective was to establish the relationship between breastfeeding, formula feeding, or mixed feeding (breastfeeding and formula) with the presence of S. mutans in a population of children under 6 months of age. Materials and Methods: The patients were recruited from the Child Care Center of Ciudad Juárez, Chihuahua, and from the pediatric dentistry postgraduate clinics of the Autonomous University of Ciudad Juárez; children exclusively fed maternally, with formula, and/or mixed were included. Those who had been fed within the previous hour were excluded. The sample was taken with a smear of the jugal groove using a sterile micro-brush. For the identification of Streptococcus mutans, a culture of Mitis Salivarius Agar (Millipore) was used. Results: 53.3% corresponded to females and 46.7% to males, 36.7% corresponded to maternal feeding, 23.3% corresponded to formula feeding, and 40% corresponded to mixed feeding. In 90% of the infants, the parents indicated that they did not perform oral hygiene. The CFU count showed that infants who were exclusively breastfed had an average of 9 × 10 CF/mL, formula-fed infants had an average of 78 × 10 CFU/mL, and those who had mixed feeding 21 × 10 CFU/mL. Conclusions: According to the results obtained, it was possible to corroborate that exclusive breastfeeding limits the colonization of Streptococcus mutans compared to those infants who receive formula or mixed feeding; these results could have a clinical impact on the dental health of infants by having a lower presence of one of the main etiological factors involved in dental caries and the type of microbiome established in the oral cavity.


Subject(s)
Breast Feeding , Milk, Human , Mouth , Streptococcus mutans , Humans , Streptococcus mutans/isolation & purification , Milk, Human/microbiology , Female , Infant , Male , Mouth/microbiology , Infant Formula/statistics & numerical data , Infant, Newborn
13.
Crit Rev Eukaryot Gene Expr ; 34(8): 59-73, 2024.
Article in English | MEDLINE | ID: mdl-39180208

ABSTRACT

The objective of this study is to assess the prevalence of antibiotic-resistant genes (ARGs) in the intestines of infants and the factors affecting their distribution. Breast milk and infant stool samples were collected from nine full-term, healthy mother-infant pairs. The bacterial distribution and various types of ARGs present in the samples were analyzed using metagenomic next-generation sequencing. Over a period spanning from 2 to 240 d after birth, a total of 273 types of ARGs were identified in both infant feces and breast milk, exhibiting a trend of increasing prevalence over time. High concentrations of representative ARG populations were identified in the intestines of infants, especially at 12-15 d after birth. These populations included APH3-Ib, tetW/N/W, mphA, and Haemophilus influenzae PBP3, and multiple ARG Escherichia coli soxS that were resistant to common clinically used aminoglycoside, tetracycline, macrolide, and beta-lactam antibiotics. Gammaproteobacteria and Bacilli, especially Enterococcus, Staphylococcus, Acinetobacter, Streptococcus, and Escherichia were among the identified ARG carriers. Maternal age and body mass index (present and before pregnancy), infant sex, maternal consumption of probiotic yogurt during pregnancy, and lactation might be substantial factors influencing the occurrence of ARG-carrying bacteria and ARG distribution in the infant feces. These results indicate that environmental factors may influence the distribution of ARG-carrying bacteria and ARGs themselves in infants during early life. Providing appropriate recommendations regarding maternal age, body mass index during pregnancy, and use of probiotic products could potentially mitigate the transmission of antibiotic-resistant microbiota and ARGs, thereby diminishing the risk of antibiotic-resistant infections and safeguarding children's health.


Subject(s)
Anti-Bacterial Agents , Feces , Milk, Human , Humans , Female , Feces/microbiology , Anti-Bacterial Agents/pharmacology , Milk, Human/microbiology , Infant , Infant, Newborn , Intestines/microbiology , Male , Drug Resistance, Bacterial/genetics , Genes, Bacterial , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/drug effects , Bacteria/genetics , Bacteria/drug effects , Drug Resistance, Microbial/genetics
14.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201270

ABSTRACT

Jaundice is a symptom of high blood bilirubin levels affecting about 80% of neonates. In neonates fed with breast milk, jaundice is particularly prevalent and severe, which is likely multifactorial. With the development of genomics and metagenomics, a deeper understanding of the neonatal gut microbiota has been achieved. We find there are accumulating evidence to indicate the importance of the gut microbiota in the mechanism of jaundice. In this paper, we present new comprehensive insight into the relationship between the microbiota and jaundice. In the new perspective, the gut is a crucial crossroad of bilirubin excretion, and bacteria colonizing the gut could play different roles in the excretion of bilirubin, including Escherichia coli as the main traffic jam causers, some Clostridium and Bacteroides strains as the traffic police, and most probiotic Bifidobacterium and Lactobacillus strains as bystanders with no effect or only a secondary indirect effect on the metabolism of bilirubin. This insight could explain why breast milk jaundice causes a longer duration of blood bilirubin and why most probiotics have limited effects on neonatal jaundice. With the encouragement of breastmilk feeding, our perspective could guide the development of new therapy methods to prevent this side effect of breastfeeding.


Subject(s)
Bilirubin , Gastrointestinal Microbiome , Jaundice, Neonatal , Probiotics , Humans , Jaundice, Neonatal/therapy , Jaundice, Neonatal/microbiology , Jaundice, Neonatal/etiology , Infant, Newborn , Bilirubin/metabolism , Bilirubin/blood , Breast Feeding , Milk, Human/microbiology , Milk, Human/metabolism
15.
mSystems ; 9(9): e0057724, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39191378

ABSTRACT

The introduction of solid foods to infants, also known as weaning, is a critical point for the development of the complex microbial community inhabiting the human colon, impacting host physiology in infancy and later in life. This research investigated in silico the impact of food-breastmilk combinations on growth and metabolite production by colonic microbes of New Zealand weaning infants using the metagenome-scale metabolic model named Microbial Community. Eighty-nine foods were individually combined with breastmilk, and the 12 combinations with the strongest influence on the microbial production of short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs) were identified. Fiber-rich and polyphenol-rich foods, like pumpkin and blackcurrant, resulted in the greatest increase in predicted fluxes of total SCFAs and individual fluxes of propionate and acetate when combined, respectively, with breastmilk. Identified foods were further combined with other foods and breastmilk, resulting in 66 multiple food-breastmilk combinations. These combinations altered in silico the impact of individual foods on the microbial production of SCFAs and BCFAs, suggesting that the interaction between the dietary compounds composing a meal is the key factor influencing colonic microbes. Blackcurrant combined with other foods and breastmilk promoted the greatest increase in the production of acetate and total SCFAs, while pork combined with other foods and breastmilk decreased the production of total BCFAs.IMPORTANCELittle is known about the influence of complementary foods on the colonic microbiome of weaning infants. Traditional in vitro and in vivo microbiome methods are limited by their resource-consuming concerns. Modeling approaches represent a promising complementary tool to provide insights into the behavior of microbial communities. This study evaluated how foods combined with other foods and human milk affect the production of short-chain fatty acids and branched-chain fatty acids by colonic microbes of weaning infants using a rapid and inexpensive in silico approach. Foods and food combinations identified here are candidates for future experimental investigations, helping to fill a crucial knowledge gap in infant nutrition.


Subject(s)
Colon , Computer Simulation , Gastrointestinal Microbiome , Milk, Human , Weaning , Humans , Milk, Human/chemistry , Milk, Human/microbiology , Milk, Human/metabolism , Gastrointestinal Microbiome/physiology , Infant , Colon/microbiology , Colon/metabolism , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis
16.
Cell Rep Med ; 5(9): 101708, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39216480

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe intestinal disease of very preterm infants with mother's own milk (MOM) providing protection, but the contribution of the MOM microbiota to NEC risk has not been explored. Here, we analyze MOM of 110 preterm infants (48 NEC, 62 control) in a cross-sectional study. Breast milk contains viable bacteria, but there is no significant difference in MOM microbiota between NEC and controls. Integrative analysis between MOM microbiota, human milk oligosaccharides (HMOs), and the infant gut microbiota shows positive correlations only between Acinetobacter in the infant gut and Acinetobacter and Staphylococcus in MOM. This study suggests that NEC protection from MOM is not modulated through the MOM microbiota. Thus, "'restoring" the MOM microbiota in donor human milk is unlikely to reduce NEC, and emphasis should instead focus on increasing fresh maternal human milk intake and researching different therapies for NEC prevention.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Premature , Milk, Human , Oligosaccharides , Humans , Milk, Human/microbiology , Milk, Human/chemistry , Enterocolitis, Necrotizing/microbiology , Oligosaccharides/metabolism , Infant, Newborn , Female , Male , Cross-Sectional Studies
17.
Am Fam Physician ; 110(2): 174-182, 2024 08.
Article in English | MEDLINE | ID: mdl-39172675

ABSTRACT

Mastitis represents a spectrum of inflammatory conditions. Lactational mastitis is the most common, with an approximate incidence of 10% in the United States, and it usually occurs in the first 3 months postpartum. Diagnosis is made clinically based on the presence of symptoms such as fever, malaise, focal breast tenderness, and overlying skin erythema or hyperpigmentation without the need for laboratory tests or imaging. However, obtaining milk cultures should be considered to guide antibiotic therapy, and ultrasonography should be performed to identify abscesses in immuno-compromised patients or those with worsening or recurrent symptoms. Because most cases of mastitis are caused by inflammation and not a true infection, a 1- to 2-day trial of conservative measures (i.e., nonsteroidal anti-inflammatory drugs, ice application, feeding the infant directly from the breast, and minimizing pumping) is often sufficient for treatment. If there is no improvement in symptoms, narrow-spectrum antibiotics may be prescribed to cover common skin flora (e.g., Staphylococcus, Streptococcus). Most patients can be treated as outpatients with oral antibiotics; however, if the condition worsens or there is a concern for sepsis, intravenous antibiotics and hospital admission may be required. Use of probiotics for treatment or prevention is not supported by good evidence. Factors that increase the risk of mastitis include overstimulation of milk production and tissue trauma from aggressive breast massage; therefore, frequent overfeeding, excessive pumping to empty the breast, heat application, and breast massage are no longer recommended because they may worsen the condition. The best prevention is a proper lactation technique, including a good infant latch, and encouraging physiologic breastfeeding rather than pumping, if possible.


Subject(s)
Anti-Bacterial Agents , Mastitis , Humans , Female , Mastitis/diagnosis , Mastitis/therapy , Anti-Bacterial Agents/therapeutic use , Breast Feeding , Milk, Human/microbiology
18.
Article in English | MEDLINE | ID: mdl-38958657

ABSTRACT

Novel Gram-positive, catalase-negative, α-haemolytic cocci were isolated from breast milk samples of healthy mothers living in Hanoi, Vietnam. The 16S rRNA gene sequences of these strains varied by 0-2 nucleotide polymorphisms. The 16S rRNA gene sequence of one strain, designated as BME SL 6.1T, showed the highest similarity to those of Streptococcus salivarius NCTC 8618T (99.4 %), Streptococcus vestibularis ATCC 49124T (99.4 %), and Streptococcus thermophilus ATCC 19258T (99.3 %) in the salivarius group. Whole genome sequencing was performed on three selected strains. Phylogeny based on 631 core genes clustered the three strains into the salivarius group, and the strains were clearly distinct from the other species in this group. The average nucleotide identity (ANI) value of strain BME SL 6.1T exhibited the highest identity with S. salivarius NCTC 8618T (88.4 %), followed by S. vestibularis ATCC 49124T (88.3 %) and S. thermophilus ATCC 19258T (87.4 %). The ANI and digital DNA-DNA hybridization values between strain BME SL 6.1T and other species were below the cut-off value (95 and 70 %, respectively), indicating that it represents a novel species of the genus Streptococcus. The strains were able to produce α-galactosidase and acid from raffinose and melibiose. Therefore, we propose to assign the strains to a new species of the genus Streptococcus as Streptococcus raffinosi sp. nov. The type strain is BME SL 6.1T (=VTCC 12812T=NBRC 116368T).


Subject(s)
Bacterial Typing Techniques , DNA, Bacterial , Milk, Human , Nucleic Acid Hybridization , Phylogeny , RNA, Ribosomal, 16S , Sequence Analysis, DNA , Streptococcus , RNA, Ribosomal, 16S/genetics , Humans , Female , DNA, Bacterial/genetics , Milk, Human/microbiology , Streptococcus/genetics , Streptococcus/isolation & purification , Streptococcus/classification , Vietnam , Whole Genome Sequencing
19.
Nutrients ; 16(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39064618

ABSTRACT

Milk is a biological fluid with a dynamic composition of micronutrients and bioactive molecules that serves as a vital nutrient source for infants. Milk composition is affected by multiple factors, including genetics, geographical location, environmental conditions, lactation phase, and maternal nutrition, and plays a key role in dictating its microbiome. This study addresses a less-explored aspect, comparing the microbial communities in human breast milk with those in mature milk from species that are used for milk consumption. Since mature animal milk is used as a supplement for both the infant (formula) and the child/adolescent, our main aim was to identify shared microbial communities in colostrum and mature human milk. Using 16S rRNA metagenomic sequencing, we focused on characterizing the milk microbiota in the Northern Greek population by identifying shared microbial communities across samples and comparing the relative abundance of prevalent genera. We analyzed ten human milk samples (from five mothers), with five collected three days postpartum (colostrum) and five collected thirty to forty days postpartum (mature milk) from corresponding mothers. To perform an interspecies comparison of human milk microbiota, we analyzed five goat and five bovine milk samples from a local dairy industry, collected fifty to seventy days after birth. Alpha diversity analysis indicated moderate diversity and stability in bovine milk, high richness in goat milk, and constrained diversity in breast milk. Beta diversity analysis revealed significant distinctions among mammalian species, emphasizing both presence/absence and abundance-based clustering. Despite noticeable differences, shared microbial components underscore fundamental aspects across all mammalian species, highlighting the presence of a core microbiota predominantly comprising the Proteobacteria, Firmicutes, and Actinobacteriota phyla. At the genus level, Acinetobacter, Gemella, and Sphingobium exhibit significant higher abundance in human milk compared to bovine and goat milk, while Pseudomonas and Atopostipes are more prevalent in animal milk. Our comparative analysis revealed differences and commonalities in the microbial communities of various mammalian milks and unraveled the existence of a common fundamental milk core microbiome. We thus revealed both species-specific and conserved microbial communities in human, bovine, and goat milk. The existence of a common core microbiome with conserved differences between colostrum and mature human milk underscores fundamental similarities in the microbiota of milk across mammalian species, which could offer valuable implications for optimizing the nutritional quality and safety of dairy products as well as supplements for infant health.


Subject(s)
Colostrum , Goats , Microbiota , Milk, Human , Milk , RNA, Ribosomal, 16S , Animals , Humans , Milk, Human/microbiology , Milk, Human/chemistry , RNA, Ribosomal, 16S/genetics , Greece , Female , Cattle , Colostrum/microbiology , Milk/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
20.
Nutrients ; 16(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999725

ABSTRACT

The correct initial colonization and establishment of the gut microbiota during the early stages of life is a key step, with long-lasting consequences throughout the entire lifespan of the individual. This process is affected by several perinatal factors; among them, feeding mode is known to have a critical role. Breastfeeding is the optimal nutrition for neonates; however, it is not always possible, especially in cases of prematurity or early pathology. In such cases, most commonly babies are fed with infant formulas in spite of the official nutritional and health international organizations' recommendation on the use of donated human milk through milk banks for these cases. However, donated human milk still does not totally match maternal milk in terms of infant growth and gut microbiota development. The present review summarizes the practices of milk banks and hospitals regarding donated human milk, its safety and quality, and the health outcomes in infants fed with donated human milk. Additionally, we explore different alternatives to customize pasteurized donated human milk with the aim of finding the perfect match between each baby and banked milk for promoting the establishment of a beneficial gut microbiota from the early stages of life.


Subject(s)
Gastrointestinal Microbiome , Infant Nutritional Physiological Phenomena , Milk Banks , Milk, Human , Humans , Milk, Human/microbiology , Infant, Newborn , Infant , Breast Feeding , Infant Formula , Female
SELECTION OF CITATIONS
SEARCH DETAIL