1
|
Reuben RC, Torres C. Integrating the milk microbiome signatures in mastitis: milk-omics and functional implications. World J Microbiol Biotechnol 2025; 41:41. [PMID: 39826029 PMCID: PMC11742929 DOI: 10.1007/s11274-024-04242-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
Mammalian milk contains a variety of complex bioactive and nutritional components and microorganisms. These microorganisms have diverse compositions and functional roles that impact host health and disease pathophysiology, especially mastitis. The advent and use of high throughput omics technologies, including metagenomics, metatranscriptomics, metaproteomics, metametabolomics, as well as culturomics in milk microbiome studies suggest strong relationships between host phenotype and milk microbiome signatures in mastitis. While single omics studies have undoubtedly contributed to our current understanding of milk microbiome and mastitis, they often provide limited information, targeting only a single biological viewpoint which is insufficient to provide system-wide information necessary for elucidating the biological footprints and molecular mechanisms driving mastitis and milk microbiome dysbiosis. Therefore, integrating a multi-omics approach in milk microbiome research could generate new knowledge, improve the current understanding of the functional and structural signatures of the milk ecosystem, and provide insights for sustainable mastitis control and microbiome management.
Collapse
Affiliation(s)
- Rine Christopher Reuben
- Biology Department, King's College, 133 North River Street, Wilkes-Barre, PA, 18711, USA.
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain.
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain
| |
Collapse
|
2
|
Asenova A, Hristova H, Ivanova S, Miteva V, Zhivkova I, Stefanova K, Moncheva P, Nedeva T, Urshev Z, Marinova-Yordanova V, Georgieva T, Tzenova M, Russinova M, Borisova T, Donchev D, Hristova P, Rasheva I. Identification and Characterization of Human Breast Milk and Infant Fecal Cultivable Lactobacilli Isolated in Bulgaria: A Pilot Study. Microorganisms 2024; 12:1839. [PMID: 39338513 PMCID: PMC11433867 DOI: 10.3390/microorganisms12091839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
During the last few decades, the main focus of numerous studies has been on the human breast milk microbiota and its influence on the infant intestinal microbiota and overall health. The presence of lactic acid bacteria in breast milk affects both the quantitative and qualitative composition of the infant gut microbiota. The aim of this study was to assess the most frequently detected cultivable rod-shaped lactobacilli, specific for breast milk of healthy Bulgarian women and fecal samples of their infants over the first month of life, in 14 mother-infant tandem pairs. Additionally, we evaluated the strain diversity among the most common isolated species. A total of 68 Gram-positive and catalase-negative strains were subjected to identification using the MALDI-TOF technique. Predominant cultivable populations belonging to the rod-shaped lactic acid bacteria have been identified as Lacticaseibacillus rhamnosus, Limosilactobacillus fermentum, Lacticaseibacillus paracasei, and Limosilactobacillus reuteri. Also, we confirmed the presence of Lactiplantibacillus plantarum and Lactobacillus gasseri. Up to 26 isolates were selected as representatives and analyzed by 16S rRNA sequencing for strain identity confirmation and a phylogenetic tree based on 16S rRNA gene sequence was constructed. Comparative analysis by four RAPD primers revealed genetic differences between newly isolated predominant L. rhamnosus strains. This pilot study provides data for the current first report concerning the investigation of the characteristic cultivable lactobacilli isolated from human breast milk and infant feces in Bulgaria.
Collapse
Affiliation(s)
- Asya Asenova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Hristiyana Hristova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Stanimira Ivanova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Viliana Miteva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Ivelina Zhivkova
- Department of Clinical Microbiology, National Center of Infectious and Parasitic Disease, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria (D.D.)
| | - Katerina Stefanova
- Agrobioinstitute Bulgarian Agriculture Academy, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria;
| | - Penka Moncheva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Trayana Nedeva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Zoltan Urshev
- LB Bulgaricum PLC, Malashevska Str. 14, 1113 Sofia, Bulgaria
| | - Victoria Marinova-Yordanova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
- Department of Bioactivity of Compouds, Centre of Competence “Sustainable Utilization of Bio Resources and Waste of Medicinal and Aromatic Plants for Innovative Bioactive Products”, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria
| | - Tzveta Georgieva
- Department of Applied Genomics and GMO, National Center of Public Health and Analyses, Academic Ivan Geshov Blvd 15, 1431 Sofia, Bulgaria;
| | - Margarita Tzenova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Maria Russinova
- Human Milk Bank, Sava Mihailov Str. 57, 1309 Sofia, Bulgaria (T.B.)
| | | | - Deyan Donchev
- Department of Clinical Microbiology, National Center of Infectious and Parasitic Disease, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria (D.D.)
- Centre of Competence “Fundamental Translational and Clinical Research in Infection and Immunity”, Yanko Sakuzov Blvd 26, 1504 Sofia, Bulgaria
| | - Petya Hristova
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| | - Iliyana Rasheva
- Department of General and Industrial Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Dragan Tsankov Blvd 8, 1164 Sofia, Bulgaria; (A.A.); (T.N.); (P.H.)
| |
Collapse
|
3
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Sáez-Fuertes L, Kapravelou G, Grases-Pintó B, Bernabeu M, Knipping K, Garssen J, Bourdet-Sicard R, Castell M, Collado MC, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Maternal Synbiotic Supplementation with B. breve M-16V and scGOS/lcFOS Shape Offspring Immune Development and Gut Microbiota at the End of Suckling. Nutrients 2024; 16:1890. [PMID: 38931246 PMCID: PMC11206815 DOI: 10.3390/nu16121890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Immune system development during gestation and suckling is significantly modulated by maternal environmental and dietary factors. Breastfeeding is widely recognized as the optimal source of nutrition for infant growth and immune maturation, and its composition can be modulated by the maternal diet. In the present work, we investigated whether oral supplementation with Bifidobacterium breve M-16V and short-chain galacto-oligosaccharide (scGOS) and long-chain fructo-oligosaccharide (lcFOS) to rat dams during gestation and lactation has an impact on the immune system and microbiota composition of the offspring at day 21 of life. On that day, blood, adipose tissue, small intestine (SI), mesenteric lymph nodes (MLN), salivary gland (SG), cecum, and spleen were collected. Synbiotic supplementation did not affect the overall body or organ growth of the pups. The gene expression of Tlr9, Muc2, IgA, and Blimp1 were upregulated in the SI, and the increase in IgA gene expression was further confirmed at the protein level in the gut wash. Synbiotic supplementation also positively impacted the microbiota composition in both the small and large intestines, resulting in higher proportions of Bifidobacterium genus, among others. In addition, there was an increase in butanoic, isobutanoic, and acetic acid concentrations in the cecum but a reduction in the small intestine. At the systemic level, synbiotic supplementation resulted in higher levels of immunoglobulin IgG2c in plasma, SG, and MLN, but it did not modify the main lymphocyte subsets in the spleen and MLN. Overall, synbiotic maternal supplementation is able to positively influence the immune system development and microbiota of the suckling offspring, particularly at the gastrointestinal level.
Collapse
Affiliation(s)
- Laura Sáez-Fuertes
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Garyfallia Kapravelou
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
| | - Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Manuel Bernabeu
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Karen Knipping
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Institute for Pharmaceutical Sciences, 3584 Utrecht, The Netherlands
| | - Johan Garssen
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Institute for Pharmaceutical Sciences, 3584 Utrecht, The Netherlands
| | | | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Center for Biomedical Research Network for the Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Carmen Collado
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Francisco José Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
5
|
Hussein F. Natural Anti-cariogenic Agents. NATURAL CONSERVATIVE DENTISTRY: AN ALTERNATIVE APPROACH TO SOLVE RESTORATIVE PROBLEMS 2024:1-40. [DOI: 10.2174/9789815223194124010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Tooth decay is primarily caused by demineralization resulting from acids
secreted by bacteria, especially Streptococcus mutans and lactobacillus, which ferment
dietary carbohydrates. This occurs in plaque biofilms, which attach to the surfaces of
the tooth and become laden with bacteria. Thus, over time, dental caries result from the
interaction of three main contributing factors: a diet containing carbohydrates, cariesproducing bacteria, as well as sensitive tooth structure. The use of an antibacterial
strategy for treating caries has evolved as a result of (1) identifying certain types of the
oral microbiome as the main cariogenic flora and (2) increasing the knowledge of the
specific ecology of these cariogenic florae. Combined with this concept, control, and
prevention of caries have been sought by reducing the number of bacteria colonizing.
Reducing bacterial populations or completely eradicating them from the oral flora
would provide an additional reason to prevent dental caries. Several undesirable sideeffects of conventional antimicrobial agents include tooth discoloration and the
emergence of bacterial resistance. These side effects stimulate the search for alternative
natural anti-microbial agents.
Collapse
Affiliation(s)
- Fatma Hussein
- Department of Operative Dentistry, Faculty of Dental Medicine for Girls, Al-Azhar University,
Cairo, Egypt
| |
Collapse
|
6
|
Ma H, Li Y, Han P, Zhang R, Yuan J, Sun Y, Li J, Chen J. Effects of Supplementing Drinking Water of Parental Pigeons with Enterococcus faecium and Bacillus subtilis on Antibody Levels and Microbiomes in Squabs. Animals (Basel) 2024; 14:178. [PMID: 38254347 PMCID: PMC10812638 DOI: 10.3390/ani14020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Enterococcus faecium (E. faecium) and Bacillus subtilis (B. subtilis) are widely used as probiotics to improve performance in animal production, but there have been few reports of their impacts on pigeon milk. In this study, twenty-four pairs of parental pigeons were randomly divided into four groups, with six replicates, and each pair feeding three squabs. The control group drank normal water. The E. faecium group, B. subtilis group, and mixed group drank water supplemented with 3 × 106 CFU/mL E. faecium, 2 × 107 CFU/mL B. subtilis, and a mixture of these two probiotics, respectively. The experiment lasted 19 days. The results demonstrated that the IgA and IgG levels were significantly higher in the milk of Group D pigeons than in the other groups. At the phylum level, Fimicutes, Actinobacteria, and Bacteroidetes were the three main phyla identified. At the genus level, Lactobacillus, Bifidobacterium, Veillonella, and Enterococcus were the four main genera identified. In conclusion, drinking water supplemented with E. faecium and B. subtilis could improve immunoglobulin levels in pigeon milk, and this could increase the ability of squabs to resist disease. E. faecium and B. subtilis could be used as probiotics in the pigeon industry.
Collapse
Affiliation(s)
- Hui Ma
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| | - Yunlei Li
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| | - Pengmin Han
- Ningxia Xiaoming Agriculture and Animal Husbandry Limited Company, Yinchuan 750000, China;
| | - Ran Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| | - Jingwei Yuan
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| | - Yanyan Sun
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| | - Jianhui Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030800, China
| | - Jilan Chen
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.M.); (Y.L.); (R.Z.); (J.Y.); (Y.S.)
| |
Collapse
|
7
|
Alemu BK, Azeze GG, Wu L, Lau SL, Wang CC, Wang Y. Effects of maternal probiotic supplementation on breast milk microbiome and infant gut microbiome and health: a systematic review and meta-analysis of randomized controlled trials. Am J Obstet Gynecol MFM 2023; 5:101148. [PMID: 37660760 DOI: 10.1016/j.ajogmf.2023.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE The early-life microbiome is formed during the perinatal period and is critical for infants' lifelong health. This is established by maternal-infant microbiome crosstalk, which is mediated by the breast milk microbiome. The milk microbiome is dependent on the maternal gut microbiome, suggesting that it could potentially be restored through oral probiotic supplements. Therefore, we conducted this systematic review and meta-analysis to summarize the effect of maternal probiotic supplements on breast milk and infant gut microbiome composition and on infant health. DATA SOURCES The PubMed, EMBASE, Web of Science, Scopus, CINAHL, and Science Direct databases were searched until December 15, 2022. STUDY ELIGIBILITY CRITERIA Randomized controlled trials following the population, intervention, comparison, and outcome (population: pregnant or lactating women; intervention: probiotics; control: placebo or follow-up; outcome: breast milk and infant gut microbiome composition and infant health) principles were included. METHODS Using a random effect model, the standard mean difference, risk difference, and risk ratio with 95% confidence interval were used to measure each outcome. All analyses were conducted using the intention-to-treat approach. Heterogeneity was evaluated using I2 statistics. RESULTS The final data set included 24 randomized controlled trials with a total of 2761 mothers and 1756 infants. The overall effect of probiotics on the beneficial bacteria detection rate in breast milk had a risk difference of 24% (95% confidence interval, 0.1-0.37; P<.001; I2=91.12%). The pooled mean beneficial and pathogenic bacteria abundance in breast milk had a standard mean difference of 1.22 log10 colony forming units/mL (95% confidence interval, 0.48-1.97; P<.001; I2=95.51%) and -1.05 log10 colony forming unites/mL (95% confidence interval, -1.99 to -0.12; P=.03; I2=96.79%), respectively. The overall abundance of beneficial bacteria in the infant gut had a standard mean difference of 0.89 log10 colony forming units/g (95% confidence interval, 0.22-1.56; P=.01; I2=95.01%). It also controlled infant weight gain (standard mean difference, -0.49 kg/equivalent age; 95% confidence interval, -0.82 to -0.17; P<.001; I2=0.00%) and decreased the occurrence of infantile colic (risk ratio, 0.30; 95% confidence interval, 0.16-0.57; P<.001; I2=0.00%). CONCLUSION Maternal probiotic supplements effectively orchestrate the breast milk and infant gut microbiome with a wide range of clinical benefits and safety. Lactobacillus, Bifidobacterium, Streptococcus thermophilus, and S. boulardii can be used as maternal supplements to promote infant health.
Collapse
Affiliation(s)
- Bekalu Kassie Alemu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Department of Midwifery, College of Medicine and Health Sciences, Debre Markos University, Ethiopia (Mr Alemu)
| | - Getnet Gedefaw Azeze
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Department of Midwifery, College Medicine and Health Sciences, Injibara University, Ethiopia (Mr Azeze)
| | - Ling Wu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang)
| | - So Ling Lau
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang)
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang); Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, Hong Kong SAR (Dr C Wang)
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR (Messers Alemu and Azeze and Drs Wu, Lau, C Wang, and Y Wang).
| |
Collapse
|
8
|
Borka Balas R, Meliț LE, Lupu A, Lupu VV, Mărginean CO. Prebiotics, Probiotics, and Synbiotics-A Research Hotspot for Pediatric Obesity. Microorganisms 2023; 11:2651. [PMID: 38004665 PMCID: PMC10672778 DOI: 10.3390/microorganisms11112651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Childhood obesity is a major public health problem worldwide with an increasing prevalence, associated not only with metabolic syndrome, insulin resistance, hypertension, dyslipidemia, and non-alcoholic fatty liver disease (NAFLD), but also with psychosocial problems. Gut microbiota is a new factor in childhood obesity, which can modulate the blood lipopolysaccharide levels, the satiety, and fat distribution, and can ensure additional calories to the host. The aim of this review was to assess the differences and the impact of the gut microbial composition on several obesity-related complications such as metabolic syndrome, NAFLD, or insulin resistance. Early dysbiosis was proven to be associated with an increased predisposition to obesity. Depending on the predominant species, the gut microbiota might have either a positive or negative impact on the development of obesity. Prebiotics, probiotics, and synbiotics were suggested to have a positive effect on improving the gut microbiota and reducing cardio-metabolic risk factors. The results of clinical trials regarding probiotic, prebiotic, and synbiotic administration in children with metabolic syndrome, NAFLD, and insulin resistance are controversial. Some of them (Lactobacillus rhamnosus bv-77, Lactobacillus salivarius, and Bifidobacterium animalis) were proven to reduce the body mass index in obese children, and also improve the blood lipid content; others (Bifidobacterium bifidum, Bifidobacterium longum, Lactobacillus acidophilus, Lacticaseibacillus rhamnosus, Enterococcus faecium, and fructo-oligosaccharides) failed in proving any effect on lipid parameters and glucose metabolism. Further studies are necessary for understanding the mechanism of the gut microbiota in childhood obesity and for developing low-cost effective strategies for its management.
Collapse
Affiliation(s)
- Reka Borka Balas
- Department of Pediatrics I, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, Gheorghe Marinescu Street, No. 38, 540136 Târgu Mureș, Romania; (R.B.B.); (C.O.M.)
| | - Lorena Elena Meliț
- Department of Pediatrics I, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, Gheorghe Marinescu Street, No. 38, 540136 Târgu Mureș, Romania; (R.B.B.); (C.O.M.)
| | - Ancuța Lupu
- Department of Pediatrics, University of Medicine and Pharmacy Gr. T. Popa Iași, Universității Street No 16, 700115 Iași, Romania; (A.L.); (V.V.L.)
| | - Vasile Valeriu Lupu
- Department of Pediatrics, University of Medicine and Pharmacy Gr. T. Popa Iași, Universității Street No 16, 700115 Iași, Romania; (A.L.); (V.V.L.)
| | - Cristina Oana Mărginean
- Department of Pediatrics I, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology, Gheorghe Marinescu Street, No. 38, 540136 Târgu Mureș, Romania; (R.B.B.); (C.O.M.)
| |
Collapse
|
9
|
Wala SJ, Ragan MV, Sajankila N, Volpe SG, Purayil N, Dumbauld Z, Besner GE. Probiotics and novel probiotic delivery systems. Semin Pediatr Surg 2023; 32:151307. [PMID: 37295299 DOI: 10.1016/j.sempedsurg.2023.151307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is an infectious and inflammatory intestinal disease that is the most common surgical emergency in the premature patient population. Although the etiology of the disease is multifactorial, intestinal dysbiosis is a hallmark of this disease. Based on this, probiotics may play a therapeutic role in NEC by introducing beneficial bacteria with immunomodulating, antimicrobial, and anti-inflammatory functions into the gastrointestinal tract. Currently, there is no Food and Drug Administration (FDA)-approved probiotic for the prevention and treatment of NEC. All probiotic clinical studies to date have administered the bacteria in their planktonic (free-living) state. This review will discuss established probiotic delivery systems including planktonic probiotics, prebiotics, and synbiotics, as well as novel probiotic delivery systems such as biofilm-based and designer probiotics. We will also shed light on whether or not probiotic efficacy is influenced by administration with breast milk. Finally, we will consider the challenges associated with developing an FDA-approved probiotic for NEC.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nitin Sajankila
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Samuel G Volpe
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nanditha Purayil
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zachary Dumbauld
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
10
|
Weerasuriya W, Saunders JE, Markel L, Ho TTB, Xu K, Lemas DJ, Groer MW, Louis-Jacques AF. Maternal gut microbiota in the postpartum Period: A Systematic review. Eur J Obstet Gynecol Reprod Biol 2023; 285:130-147. [PMID: 37116306 PMCID: PMC10320739 DOI: 10.1016/j.ejogrb.2023.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/26/2023] [Accepted: 03/31/2023] [Indexed: 04/03/2023]
Abstract
Studies have demonstrated the importance of the gut microbiota during pregnancy, and there is emerging literature on the postpartum maternal gut microbiota. The primary objective of this paper was to synthesize the literature on the postpartum gut microbiome composition and diversity measured in stool samples from healthy mothers of predominantly term infants. The secondary objectives were (1) to identify biological and environmental factors that influence postpartum maternal gut microbiota and (2) to assess health conditions and clinical intermediate measures associated with postpartum gut microbiota changes in all mothers. Electronic searches were conducted November 9, 2020 and updated July 25, 2021 without publication time limits on PubMed, Embase, CINHAL, Scopus, Cochrane Library, BioArchives, and OpenGrey.eu. Primary research on maternal gut microbiota in the postpartum (up to one year after childbirth) were eligible. Postpartum gut microbiota comparisons to pregnancy or non-pregnancy gut microbiota were of interest, therefore, studies examining these in addition to the postpartum were included. Studies were excluded if they were only conducted in animals, infants, pregnancy, or microbiome of other body locations (e.g., vaginal). Data extraction of microbial composition and diversity were completed and synthesized narratively. Studies were assessed for risk of bias. A total of 2512 articles were screened after deduplication and 27 were included in this review. Of the 27 included studies, 22 addressed the primary objective. Firmicutes was the predominant phylum in the early (<6 weeks) and late postpartum (6 weeks to 1 year). In early postpartum, Bacteroides was the predominant genus. Findings from longitudinal assessments of alpha and beta diversity from the early to the late postpartum varied. Nineteen of the 27 studies assessed biological and environmental factors influencing the postpartum gut microbial profile changes. Timing of delivery, probiotic supplementation, triclosan exposure, and certain diets influenced the postpartum gut microbiota. Regarding health conditions and intermediate clinical measures assessed in 8 studies; inflammatory bowel disease, postpartum depression, early-onset preeclampsia, gestational diabetes, excessive gestational weight gain, and anthropometric measures such as body mass index and waist-to-hip ratio were related to gut microbiota changes. There is limited data on the maternal postpartum gut microbiota and how it influences maternal health. We need to understand the postpartum maternal gut microbiome, establish how it differs from non-pregnancy and pregnancy states, and determine biological and environmental influencers. Future research of the gut microbiome's significance for the birthing parent in the postpartum could lead to a new understanding of how to improve maternal short and long-term health.
Collapse
Affiliation(s)
- Wasana Weerasuriya
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida (UF), Gainesville, FL, USA
| | - Julia E Saunders
- Morsani College of Medicine, University of South Florida (USF), Tampa, FL, USA
| | - Lilla Markel
- Morsani College of Medicine, University of South Florida (USF), Tampa, FL, USA
| | - Thao T B Ho
- Department of Pediatrics, Morsani College of Medicine, USF, Tampa, FL, USA
| | - Ke Xu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, UF, Gainesville, FL, USA
| | - Dominick J Lemas
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida (UF), Gainesville, FL, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, UF, Gainesville, FL, USA
| | - Maureen W Groer
- College of Nursing, University of Tennessee, Knoxville, TN, USA
| | - Adetola F Louis-Jacques
- Department of Obstetrics and Gynecology, College of Medicine, University of Florida (UF), Gainesville, FL, USA.
| |
Collapse
|
11
|
Mills S, Yang B, Smith GJ, Stanton C, Ross RP. Efficacy of Bifidobacterium longum alone or in multi-strain probiotic formulations during early life and beyond. Gut Microbes 2023; 15:2186098. [PMID: 36896934 PMCID: PMC10012958 DOI: 10.1080/19490976.2023.2186098] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
The significance of Bifidobacterium to human health can be appreciated from its early colonization of the neonatal gut, where Bifidobacterium longum represents the most abundant species. While its relative abundance declines with age, it is further reduced in several diseases. Research into the beneficial properties of B. longum has unveiled a range of mechanisms, including the production of bioactive molecules, such as short-chain fatty acids, polysaccharides, and serine protease inhibitors. From its intestinal niche, B. longum can have far-reaching effects in the body influencing immune responses in the lungs and even skin, as well as influencing brain activity. In this review, we present the biological and clinical impacts of this species on a range of human conditions beginning in neonatal life and beyond. The available scientific evidence reveals a strong rationale for continued research and further clinical trials that investigate the ability of B. longum to treat or prevent a range of diseases across the human lifespan.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Co Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Du Y, Qiu Q, Cheng J, Huang Z, Xie R, Wang L, Wang X, Han Z, Jin G. Comparative study on the microbiota of colostrum and nipple skin from lactating mothers separated from their newborn at birth in China. Front Microbiol 2022; 13:932495. [PMID: 36262322 PMCID: PMC9574262 DOI: 10.3389/fmicb.2022.932495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing studies have found breast milk (BM) contains its own microbiota. However, the route through which microbes enter the BM is still unclear. In order to verify the entero-mammary pathway of BM, we designed a rigorous study that prevented oral bacteria from contaminating the breast and nipple skin (NS) during baby nursing. Thirty-one healthy, postpartum mothers living in southern China who were immediately separated from their newborn after delivery were enrolled in this study. Using an aseptic protocol for sampling, sterile water was used to wash the NS and was then collected. Then the first drop of BM was discarded and colostrum was collected manually. Amplicon sequencing was performed targeting the V3–V4 region of the bacterial 16S rRNA gene, and the differences between the microbiota of the colostrum and NS were analyzed. Additionally, the effects of environmental factors, such as the delivery mode and intrapartum antibiotic exposure, on the diversity of the colostrum microbiota were also analyzed. We found significant differences in the α diversity and richness between the BM and NS as evidenced by richness, Chao1, and Simpson indices. There were 170 operational taxonomic units (OTUs) shared by colostrum and NS, while 111 and 87 OTUs were unique, respectively, as well as a clear distinction in OTUs was observed by unifrac binary analysis between them. Linear discriminant analysis effect size analysis found that anaerobes, such as Bifidobacterium and Pantoea at the genus level and enterobacteria including Enterobacteriaceae at the family level, were predominant in the colostrum, while the predominant bacteria on the NS were Bacteroides, Staphylococcus, and Parabacteroides at the genus level. BM is colonized by bacteria prior to baby suckling, and the diversity of the colostrum microbiota differs from that of the NS. The predominant microbiota taxa in BM indicated that they were likely to be transferred to the breast through the intestinal tract. Our study provides direct evidence for the revolutionary active migration hypothesis. Additionally, factors like intrapartum antibiotic exposure did not significantly affect the diversity of the microbiota in the BM. Therefore, it is suggested that mothers continue to provide BM for their newborns during separation.
Collapse
Affiliation(s)
- Yanli Du
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Qing Qiu
- Department of Women Health Care, Shenzhen Luohu Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Jing Cheng
- Department of Obstetrics, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhili Huang
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Ruixia Xie
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
| | - Lu Wang
- Delivery Center, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Xiangyu Wang
- Shenzhen Second People’s Hospital, Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Xiangyu Wang,
| | - Zongli Han
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Zongli Han,
| | - Gang Jin
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen, China
- Gang Jin,
| |
Collapse
|
13
|
Tunay RT, Kök Taş T. Verticle transmission of unique bacterial strains from mother to infant via consuming natural kefir. Int Dairy J 2022. [DOI: 10.1016/j.idairyj.2021.105251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
In silico, in vitro and in vivo safety evaluation of Limosilactobacillus reuteri strains ATCC PTA-126787 & ATCC PTA-126788 for potential probiotic applications. PLoS One 2022; 17:e0262663. [PMID: 35081129 PMCID: PMC8791467 DOI: 10.1371/journal.pone.0262663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 01/02/2022] [Indexed: 11/19/2022] Open
Abstract
The last two decades have witnessed a tremendous growth in probiotics and in the numbers of publications on their potential health benefits. Owing to their distinguishing beneficial effects and long history of safe use, species belonging to the Lactobacillus genus are among the most widely used probiotic species in human food and dietary supplements and are finding increased use in animal feed. Here, we isolated, identified, and evaluated the safety of two novel Limosilactobacillus reuteri (L. reuteri) isolates, ATCC PTA-126787 & ATCC PTA-126788. More specifically, we sequenced the genomes of these two L. reuteri strains using the PacBio sequencing platform. Using a combination of biochemical and genetic methods, we identified the two strains as belonging to L. reuteri species. Detailed in silico analyses showed that the two strains do not encode for any known genetic sequences of concern for human or animal health. In vitro assays confirmed that the strains are susceptible to clinically relevant antibiotics and do not produce potentially harmful by-products such as biogenic amines. In vitro bile and acid tolerance studies demonstrated that the two strains have similar survival profiles as the commercial L. reuteri probiotic strain DSM 17938. Most importantly, daily administration of the two probiotic strains to broiler chickens in drinking water for 26 days did not induce any adverse effect, clinical disease, or histopathological lesions, supporting the safety of the strains in an in vivo avian model. All together, these data provide in silico, in vitro and in vivo evidence of the safety of the two novel candidates for potential probiotic applications in humans as well as animals.
Collapse
|
15
|
Duale A, Singh P, Al Khodor S. Breast Milk: A Meal Worth Having. Front Nutr 2022; 8:800927. [PMID: 35155521 PMCID: PMC8826470 DOI: 10.3389/fnut.2021.800927] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
A mother is gifted with breast milk, the natural source of nutrition for her infant. In addition to the wealth of macro and micro-nutrients, human milk also contains many microorganisms, few of which originate from the mother, while others are acquired from the mouth of the infant and the surroundings. Among these microbes, the most commonly residing bacteria are Staphylococci, Streptococci, Lactobacilli and Bifidobacteria. These microorganisms initiate and help the development of the milk microbiota as well as the microbiota of the gastrointestinal tract in infants, and contribute to developing immune regulatory factors such as cytokines, growth factors, lactoferrin among others. These factors play an important role in reducing the risk of developing chronic diseases like type 2 diabetes, asthma and others later in life. In this review, we will summarize the known benefits of breastfeeding and highlight the role of the breast milk microbiota and its cross-talk with the immune system in breastfed babies during the early years of life.
Collapse
Affiliation(s)
- Anoud Duale
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Parul Singh
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Ar-Rayyan, Qatar
| | - Souhaila Al Khodor
- Division of Maternal and Child Health, Department of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
- *Correspondence: Souhaila Al Khodor
| |
Collapse
|
16
|
de Andrade PDSMA, Maria e Silva J, Carregaro V, Sacramento LA, Roberti LR, Aragon DC, Carmona F, Roxo-Junior P. Efficacy of Probiotics in Children and Adolescents With Atopic Dermatitis: A Randomized, Double-Blind, Placebo-Controlled Study. Front Nutr 2022; 8:833666. [PMID: 35155534 PMCID: PMC8826069 DOI: 10.3389/fnut.2021.833666] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 12/31/2021] [Indexed: 12/26/2022] Open
Abstract
ObjectiveTo evaluate the clinical efficacy of a mixture of probiotics (Lactobacillus and Bifidobacterium) in children and adolescents with atopic dermatitis (AD) and the effects on sensitization, inflammation, and immunological tolerance.MethodsIn this double-blind, randomized, placebo-controlled clinical trial, we enrolled 60 patients aged between 6 months and 19 years with mild, moderate, or severe AD, according to the criteria proposed by Hanifin and Rajka. Patients were stratified to receive one gram per day of probiotics or placebo for 6 months. The primary outcome was a decrease in SCORing Atopic Dermatitis (SCORAD). Secondary outcomes were to assess the role of probiotics on the use of topical and oral medicines (standard treatment), serum IgE levels, skin prick test (SPT), and tolerogenic and inflammatory cytokines. Background therapy was maintained.ResultsForty patients completed the study (24 probiotics, 16 placebo). After treatment for six months, the clinical response was significantly better in the probiotics group; the SCORAD decreased [mean difference (MD) 27.69 percentage points; 95% confidence interval (CI), 2.44–52.94], even after adjustment for co-variables (MD 32.33 percentage points; 95%CI, 5.52–59.13), especially from the third month of treatment on. The reduction of the SCORAD in probiotic group persisted for three more months after the treatment had been discontinued, even after adjustment for co-variables (MD 14.24 percentage points; 95%CI, 0.78–27.70). Patients in the probiotics group required topical immunosuppressant less frequently at 6 and 9 months. No significant changes were found for IgE levels, SPT and cytokines.ConclusionsChildren and adolescents with AD presented a significant clinical response after 6 months with a mixture of probiotics (Lactobacillus rhamnosus, Lactobacillus acidophilus, Lactobacillus paracasei, and Bifidobacterium lactis. However, this clinical benefit is related to treatment duration. Probiotics should be considered as an adjuvant treatment for AD.
Collapse
Affiliation(s)
| | - Jorgete Maria e Silva
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Vanessa Carregaro
- Department of Immunology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, São Paulo, Brazil
| | - Laís Amorim Sacramento
- Department of Immunology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, São Paulo, Brazil
| | | | - Davi Casale Aragon
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Fabio Carmona
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Pérsio Roxo-Junior
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- *Correspondence: Pérsio Roxo-Junior
| |
Collapse
|
17
|
Prenatal versus Postnatal Initial Colonization of Healthy Neonates' Colon Ecosystem by the Enterobacterium Escherichia coli. Microbiol Spectr 2021; 9:e0037921. [PMID: 34817225 PMCID: PMC8612161 DOI: 10.1128/spectrum.00379-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The human colon is a microbial ecosystem whose initial bacterial colonization in neonates is an important step in establishing a beneficial microbiota for the body's health. This study investigated the occurrence of viable culturable Escherichia coli in first-day meconium versus subsequent days' stool to explore the prenatal versus postnatal initial colonization of the colon by E. coli in healthy neonates. E. coli occurrence was investigated on eosin-methylene blue (EMB) agar, followed by morphological and biochemical characterizations and phylogenetic analysis of 16S rRNA-encoding gene sequences. Viable culturable E. coli was not detected in meconium of healthy male or female neonates delivered either vaginally or by cesarean section. Neonates delivered surgically also showed no E. coli colonization on the second and third days, confirming postnatal colonization of the colon by this enterobacterium. E. coli's initial colonization in the colon of neonates delivered vaginally occurred on the second day, which can be attributed to inoculation from the vaginal canal during delivery and, in comparison to the colonization in neonates delivered surgically, leads to the inference that the bacterium is not originally found in meconium. This study suggests no viability of the meconium microbiome in healthy neonates, possibly due to antimicrobial action in the prenatal colon's meconium protecting babies' gut from infection during delivery. IMPORTANCE The results of this study suggest that the initial postnatal colonization of neonates' colon by beneficial bacteria is a naturally controlled process in which the prenatal colon's meconium might play a role in protecting against infection of the babies' gut during delivery.
Collapse
|
18
|
Zeinali LI, Giuliano S, Lakshminrusimha S, Underwood MA. Intestinal Dysbiosis in the Infant and the Future of Lacto-Engineering to Shape the Developing Intestinal Microbiome. Clin Ther 2021; 44:193-214.e1. [PMID: 34922744 DOI: 10.1016/j.clinthera.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE The goal of this study was to review the role of human milk in shaping the infant intestinal microbiota and the potential of human milk bioactive molecules to reverse trends of increasing intestinal dysbiosis and dysbiosis-associated diseases. METHODS This narrative review was based on recent and historic literature. FINDINGS Human milk immunoglobulins, oligosaccharides, lactoferrin, lysozyme, milk fat globule membranes, and bile salt-stimulating lipase are complex multifunctional bioactive molecules that, among other important functions, shape the composition of the infant intestinal microbiota. IMPLICATIONS The co-evolution of human milk components and human milk-consuming commensal anaerobes many thousands of years ago resulted in a stable low-diversity infant microbiota. Over the past century, the introduction of antibiotics and modern hygiene practices plus changes in the care of newborns have led to significant alterations in the intestinal microbiota, with associated increases in risk of dysbiosis-associated disease. A better understanding of mechanisms by which human milk shapes the intestinal microbiota of the infant during a vulnerable period of development of the immune system is needed to alter the current trajectory and decrease intestinal dysbiosis and associated diseases.
Collapse
Affiliation(s)
- Lida I Zeinali
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | | | | | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
19
|
Tougas SR, Lodha N, Vandermeer B, Lorenzetti DL, Tarr PI, Tarr GAM, Chui L, Vanderkooi OG, Freedman SB. Prevalence of Detection of Clostridioides difficile Among Asymptomatic Children: A Systematic Review and Meta-analysis. JAMA Pediatr 2021; 175:e212328. [PMID: 34338715 PMCID: PMC8329794 DOI: 10.1001/jamapediatrics.2021.2328] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE Detection of Clostridioides difficile has frequently been described in asymptomatic infants and children, but accurate estimates across the age spectrum are unavailable. OBJECTIVE To assess the prevalence of C difficile detection among asymptomatic children across the age spectrum. DATA SOURCES This systematic review and meta-analysis included a search of the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, CINAHL, Scopus, and Web of Science for articles published from January 1, 1990, to December 31, 2020. Search terms included Clostridium difficile, Peptoclostridium difficile, Clostridioides difficile, CDF OR CDI OR c diff OR c difficile, Clostridium infections OR cd positive diarrhea OR cd positive diarrhea OR Clostridium difficile OR Peptoclostridium difficile OR pseudomembranous colitis OR pseudomembranous enterocolitis, enterocolitis, and pseudomembranous. These were combined with the following terms: bacterial colonization and colonization OR colonized OR colonizing OR epidemiology OR prevalence OR seroprevalence. STUDY SELECTION Studies were screened independently by 2 authors. Studies were included if they reported testing for C difficile among asymptomatic children (ie, children without diarrhea) younger than 18 years. DATA EXTRACTION AND SYNTHESIS Data were extracted independently and in duplicate by 2 reviewers. Preferred Reporting Items for a Systematic Review and Meta-analysis (PRISMA) guidelines were used. Data were pooled using a random-effects model. MAIN OUTCOMES AND MEASURES The primary outcome was prevalence of C difficile detection among asymptomatic children. Secondary outcomes included prevalence of toxigenic vs nontoxigenic strains of C difficile and prevalence of C difficile detection stratified by geographic region, income status, testing method, and year of testing. RESULTS A total of 95 studies with 19 186 participants were included. Rates of detection of toxigenic or nontoxigenic C difficile were greatest among infants aged 6 to 12 months (41%; 95% CI, 32%-50%) and decreased to 12% (95% CI, 7%-18%) among children aged 5 to 18 years. The prevalence of toxigenic C difficile colonization was lower, peaking at 14% (95% CI, 8%-21%) among infants aged 6 to 12 months and decreasing to 6% (95% CI, 2%-11%) among children older than 5 years. Although prevalence differed by geographic region (ie, North and South America vs Europe: β, -0.151, P = .001; North and South America vs Western Pacific: β, 0.136, P = .007), there was no difference by testing method (ie, culture vs polymerase chain reaction: β, 0.069, P = .052; culture vs enzyme immunoassay: β, -0.178, P = .051), income class (low-middle income vs high income: β, -0.144, P = .23; upper-middle vs high income: β, -0.020, P = .64), or period (before 1990 vs 2010-2020: β, -0.125, P = .19; 1990-1999 vs 2010-2020: β, -0.037, P = .42; 2000-2009 vs 2010-2020: β, -0.006, P = .86). CONCLUSIONS AND RELEVANCE In this systematic review and meta-analysis, C difficile colonization rates among children were greatest at 6 to 12 months of age and decreased thereafter. These estimates may provide context for interpreting C difficile test results among young children.
Collapse
Affiliation(s)
- Sarah R Tougas
- Cumming School of Medicine, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Nidhi Lodha
- Cumming School of Medicine, Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Ben Vandermeer
- Alberta Research Centre for Health Evidence, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Diane L Lorenzetti
- Department of Community Health Sciences and the Health Sciences Library, University of Calgary, Calgary, Alberta, Canada
| | - Phillip I Tarr
- Department of Pediatrics, Washington University in St Louis, St Louis, Missouri.,Department of Molecular Microbiology, Washington University in St Louis, St Louis, Missouri
| | - Gillian A M Tarr
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis
| | - Linda Chui
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Otto G Vanderkooi
- Alberta Children's Hospital, Division of Pediatric Infectious Disease, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Microbiology, Immunology and Infectious Diseases, Community Health Sciences, and Pathology & Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephen B Freedman
- Alberta Children's Hospital, Divisions of Pediatric Emergency Medicine and Gastroenterology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Department of Emergency Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Guo J, Han X, Huang W, You Y, Zhan J. Gut dysbiosis during early life: causes, health outcomes, and amelioration via dietary intervention. Crit Rev Food Sci Nutr 2021; 62:7199-7221. [PMID: 33909528 DOI: 10.1080/10408398.2021.1912706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The colonization and maturation of gut microbiota (GM) is a delicate and precise process, which continues to influence not only infancy and childhood but also adulthood health by affecting immunity. However, many perinatal factors, including gestational age, delivery mode, antibiotic administration, feeding mode, and environmental and maternal factors, can disturb this well-designed process, increasing the morbidity of various gut dysbiosis-related diseases, such as type-1-diabetes, allergies, necrotizing enterocolitis, and obesity. In this review, we discussed the early-life colonization and maturation of the GM, factors influencing this process, and diseases related to the disruption of this process. Moreover, we focused on discussing dietary interventions, including probiotics, oligosaccharides, nutritional supplementation, and exclusive enteral nutrition, in ameliorating early-life dysbiosis and diseases related to it. Furthermore, possible mechanisms, and shortcomings, as well as potential solutions to the drawbacks of dietary interventions, were also discussed.
Collapse
Affiliation(s)
- Jielong Guo
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Xue Han
- Peking University School of Basic Medical Science, Peking University Health Science Centre, Beijing, China
| | - Weidong Huang
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Yilin You
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Jicheng Zhan
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| |
Collapse
|
21
|
Lactobacillus reuteri Colonisation of Extremely Preterm Infants in a Randomised Placebo-Controlled Trial. Microorganisms 2021; 9:microorganisms9050915. [PMID: 33923278 PMCID: PMC8190634 DOI: 10.3390/microorganisms9050915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022] Open
Abstract
Lactobacillus reuteri DSM 17938 supplementation reduces morbidities in very low birth weight infants (<1500 g), while the effect on extremely low birth weight infants (ELBW, <1000 g) is still questioned. In a randomised placebo-controlled trial (ClinicalTrials.gov ID NCT01603368), head growth, but not feeding tolerance or morbidities, improved in L. reuteri-supplemented preterm ELBW infants. Here, we investigate colonisation with the probiotic strain in preterm ELBW infants who received L. reuteri DSM 17938 or a placebo from birth to postmenstrual week (PMW) 36. Quantitative PCR was used on 582 faecal DNA samples collected from 132 ELBW infants at one, two, three, and four weeks, at PMW 36, and at two years of age. Human milk oligosaccharides were measured in 31 milk samples at two weeks postpartum. At least 86% of the ELBW infants in the L. reuteri group were colonised with the probiotic strain during the neonatal period, despite low gestational age, high antibiotic pressure, and independent of infant feeding mode. Higher concentrations of lacto-N-tetraose, sialyl-lacto-N-neotetraose c, and 6′-sialyllactose in mother’s milk weakly correlated with lower L. reuteri abundance. Within the L. reuteri group, higher L. reuteri abundance weakly correlated with a shorter time to reach full enteral feeding. Female sex and L. reuteri colonisation improved head growth from birth to four weeks of age. In conclusion, L. reuteri DSM 17938 supplementation leads to successful colonisation in ELBW infants.
Collapse
|
22
|
Zaidi AZ, Moore SE, Okala SG. Impact of Maternal Nutritional Supplementation during Pregnancy and Lactation on the Infant Gut or Breastmilk Microbiota: A Systematic Review. Nutrients 2021; 13:nu13041137. [PMID: 33808265 PMCID: PMC8067242 DOI: 10.3390/nu13041137] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
Recent evidence indicates that maternal dietary intake, including dietary supplements, during pregnancy and lactation may alter the infant gut or breastmilk microbiota, with implications for health outcomes in both the mother and infant. To review the effects of maternal nutritional supplementation during pregnancy and lactation on the infant gut or breastmilk microbiota a systematic literature search was conducted. A total of 967 studies published until February 2020 were found, 31 were eligible and 29 randomized control trials were included in the qualitative synthesis. There were 23 studies that investigated the effects of probiotic supplementation, with the remaining studies investigating vitamin D, prebiotics or lipid-based nutrient supplements (LNS). The effects of maternal nutritional supplementation on the infant gut microbiota or breastmilk microbiota were examined in 21 and 12 studies, respectively. Maternal probiotic supplementation during pregnancy and lactation generally resulted in the probiotic colonization of the infant gut microbiota, and although most studies also reported alterations in the infant gut bacterial loads, there was limited evidence of effects on bacterial diversity. The data available show that maternal probiotic supplementation during pregnancy or lactation results in probiotic colonization of the breastmilk microbiota. There were no observed effects between probiotic supplementation and breastmilk bacterial counts of healthy women, however, administration of Lactobacillus probiotic to nursing women affected by mastitis was associated with significant reductions in breastmilk Staphylococcal loads. Maternal LNS supplementation during pregnancy and lactation increased bacterial diversity in the infant gut, whilst vitamin D and prebiotic supplementation did not alter either infant gut bacterial diversity or counts. Heterogeneity in study design precludes any firm conclusions on the effects of maternal nutritional supplementation during pregnancy and lactation on the infant gut or breastmilk microbiota, warranting further research.
Collapse
Affiliation(s)
- Aneesa Z. Zaidi
- Medical School, St George’s University of London, London SW17 0RE, UK;
| | - Sophie E. Moore
- Department of Women and Children’s Health, King’s College London, London SE1 7EH, UK;
- Correspondence: ; Tel.: +44-020-7188-3639
| | - Sandra G. Okala
- Department of Women and Children’s Health, King’s College London, London SE1 7EH, UK;
| |
Collapse
|
23
|
Boudry G, Charton E, Le Huerou-Luron I, Ferret-Bernard S, Le Gall S, Even S, Blat S. The Relationship Between Breast Milk Components and the Infant Gut Microbiota. Front Nutr 2021; 8:629740. [PMID: 33829032 PMCID: PMC8019723 DOI: 10.3389/fnut.2021.629740] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
The assembly of the newborn's gut microbiota during the first months of life is an orchestrated process resulting in specialized microbial ecosystems in the different gut compartments. This process is highly dependent upon environmental factors, and many evidences suggest that early bacterial gut colonization has long-term consequences on host digestive and immune homeostasis but also metabolism and behavior. The early life period is therefore a "window of opportunity" to program health through microbiota modulation. However, the implementation of this promising strategy requires an in-depth understanding of the mechanisms governing gut microbiota assembly. Breastfeeding has been associated with a healthy microbiota in infants. Human milk is a complex food matrix, with numerous components that potentially influence the infant microbiota composition, either by enhancing specific bacteria growth or by limiting the growth of others. The objective of this review is to describe human milk composition and to discuss the established or purported roles of human milk components upon gut microbiota establishment. Finally, the impact of maternal diet on human milk composition is reviewed to assess how maternal diet could be a simple and efficient approach to shape the infant gut microbiota.
Collapse
Affiliation(s)
- Gaëlle Boudry
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | - Elise Charton
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- UMR STLO INRAE, Institut Agro, Rennes, France
| | | | | | - Sophie Le Gall
- INRAE, UR BIA, Nantes, France
- INRAE, BIBS facility, Nantes, France
| | | | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| |
Collapse
|
24
|
Afrin S, Akter S, Begum S, Hossain MN. The Prospects of Lactobacillus oris as a Potential Probiotic With Cholesterol-Reducing Property From Mother's Milk. Front Nutr 2021; 8:619506. [PMID: 33748173 PMCID: PMC7969506 DOI: 10.3389/fnut.2021.619506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
This experiment was conducted to characterize potential Lactobacillus spp. isolated from mother's milk and infant feces to obtain new and specific probiotic strains. In this study, seven ascendant strains were identified as Lactobacillus spp. based on their morphological characteristics and biochemical properties. Among them, only one (C-1) isolate was identified as Lactobacillus oris through BioLogTM identification. The study further investigated the isolate through probiotic potentiality tests such as pH and bile tolerance, NaCl tolerance test, gastric juice tolerance, antioxidant activity, resistance to hydrogen, reduction of sodium nitrate, antimicrobial activity, and antibiotic susceptibility test. The result showed that the strain is a potential probiotic based on probiotic capability. The identified strain was most acid-tolerant and retained around 80% viability for up to 4 h at pH 1.0 and 2.0. The isolate showed tolerance against up to 1.50% bile concentration and gastric juice and was able to grow 1-6% NaCl concentrations. Lactobacillus oris showed resistance to most antibiotics as well as antagonistic activity against the tested pathogen, good antioxidant properties, reduction of sodium nitrate and H2O2. The isolate exhibited good intestinal epithelial adhesion properties, and SDS page was performed for secreted protein analysis. Moreover, the strain showed promising cholesterol-lowering properties based on the cholesterol level. This present result indicates that L. oris has superior probiotic properties and can be regarded as a potential probiotic candidate.
Collapse
Affiliation(s)
- Sadia Afrin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka, Bangladesh
| | - Suraiya Akter
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Shamima Begum
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Md Nur Hossain
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka, Bangladesh
| |
Collapse
|
25
|
Shin DY, Park J, Yi DY. Comprehensive Analysis of the Effect of Probiotic Intake by the Mother on Human Breast Milk and Infant Fecal Microbiota. J Korean Med Sci 2021; 36:e58. [PMID: 33650336 PMCID: PMC7921370 DOI: 10.3346/jkms.2021.36.e58] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Human breast milk (HBM) contains optimal nutrients for infant growth. Probiotics are used to prevent disease and, when taken by the mother, they may affect infant microbiome as well as HBM. However, few studies have specifically investigated the effect of probiotic intake by the mother on HBM and infant microbiota at genus/species level. Therefore, we present a comprehensive analysis of paired HBM and infant feces (IF) microbiome samples before and after probiotic intake by HBM-producing mothers. METHODS Lactating mothers were administered with Lactobacillus rhamnosus (n = 9) or Saccharomyces boulardii capsules (n = 9), for 2 months; or no probiotic (n = 7). Paired HBM and IF samples were collected before and after treatment and analyzed by next-generation sequencing. RESULTS Forty-three HBM and 49 IF samples were collected and sequenced. Overall, in 43 HBM samples, 1,190 microbial species belonging to 684 genera, 245 families, 117 orders, and 56 classes were detected. In 49 IF samples, 372 microbial species belonging to 195 genera, 79 families, 42 orders, and 18 classes were identified. Eight of 20 most abundant genera in both HBM and IF samples overlapped: Streptococcus (14.42%), Lactobacillus, Staphylococcus, and Veillonella, which were highly abundant in the HBM samples; and Bifidobacterium (27.397%), Bacteroides, and Faecalibacterium, which were highly abundant in the IF samples. Several major bacterial genera and species were detected in the HBM and IF samples after probiotic treatment, illustrating complex changes in the microbiomes upon treatment. CONCLUSION This is the first Korean microbiome study in which the effect of different probiotic intake by the mother on the microbiota in HBM and IF samples was investigated. This study provides a cornerstone to further the understanding of the effect of probiotics on the mother and infant microbiomes.
Collapse
Affiliation(s)
- Do Young Shin
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | | | - Dae Yong Yi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
- Department of Pediatrics, College of Medicine, Chung-Ang University, Seoul, Korea.
| |
Collapse
|
26
|
The Gut‒Breast Axis: Programming Health for Life. Nutrients 2021; 13:nu13020606. [PMID: 33673254 PMCID: PMC7917897 DOI: 10.3390/nu13020606] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The gut is a pivotal organ in health and disease. The events that take place in the gut during early life contribute to the programming, shaping and tuning of distant organs, having lifelong consequences. In this context, the maternal gut plays a quintessence in programming the mammary gland to face the nutritional, microbiological, immunological, and neuroendocrine requirements of the growing infant. Subsequently, human colostrum and milk provides the infant with an impressive array of nutrients and bioactive components, including microbes, immune cells, and stem cells. Therefore, the axis linking the maternal gut, the breast, and the infant gut seems crucial for a correct infant growth and development. The aim of this article is not to perform a systematic review of the human milk components but to provide an insight of their extremely complex interactions, which render human milk a unique functional food and explain why this biological fluid still truly remains as a scientific enigma.
Collapse
|
27
|
Moossavi S, Fontes ME, Rossi L, Fusch G, Surette MG, Azad MB. Capturing the diversity of the human milk microbiota through culture-enriched molecular profiling: a feasibility study. FEMS Microbiol Lett 2021; 368:6070652. [PMID: 33417698 DOI: 10.1093/femsle/fnab001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
Previous human milk studies have confirmed the existence of a highly diverse bacterial community using culture-independent and targeted culture-dependent techniques. However, culture-enriched molecular profiling of milk microbiota has not been done. Additionally, the impact of storage conditions and milk fractionation on microbiota composition is not understood. In this feasibility study, we optimized and applied culture-enriched molecular profiling to study culturable milk microbiota in eight milk samples collected from mothers of infants admitted to a neonatal intensive care unit. Fresh samples were immediately plated or stored at -80°C for 2 weeks (short-term frozen). Long-term samples were stored at -20°C for >6 months. Samples were cultured using 10 different culture media and incubated both aerobically and anaerobically. We successfully isolated major milk bacteria, including Streptococcus, Staphylococcus and Bifidobacterium, from fresh milk samples, but were unable to culture any bacteria from the long-term frozen samples. Short-term freezing shifted the composition of viable milk bacteria from the original composition in fresh samples. Nevertheless, the inter-individual variability of milk microbiota composition was observed even after short-term storage. There was no major difference in the overall milk microbiota composition between milk fractions in this feasibility study. This is among the first studies on culture-enriched molecular profiling of the milk microbiota demonstrating the effect of storage and fractionation on milk microbiota composition.
Collapse
Affiliation(s)
- Shirin Moossavi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada.,Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran 14117-13135, Iran.,Microbiome and Microbial Ecology Interest Group (MMEIG), Universal Scientific Education and Research Network (USERN), Calgary, AB T2N 4Z1, Canada
| | - Michelle E Fontes
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Laura Rossi
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gerhard Fusch
- Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Michael G Surette
- Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada.,Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| |
Collapse
|
28
|
Grech A, Collins CE, Holmes A, Lal R, Duncanson K, Taylor R, Gordon A. Maternal exposures and the infant gut microbiome: a systematic review with meta-analysis. Gut Microbes 2021; 13:1-30. [PMID: 33978558 PMCID: PMC8276657 DOI: 10.1080/19490976.2021.1897210] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 02/04/2023] Open
Abstract
Early life, including the establishment of the intestinal microbiome, represents a critical window of growth and development. Postnatal factors affecting the microbiome, including mode of delivery, feeding type, and antibiotic exposure have been widely investigated, but questions remain regarding the influence of exposures in utero on infant gut microbiome assembly. This systematic review aimed to synthesize evidence on exposures before birth, which affect the early intestinal microbiome. Five databases were searched in August 2019 for studies exploring pre-pregnancy or pregnancy 'exposure' data in relation to the infant microbiome. Of 1,441 publications identified, 76 were included. Factors reported influencing microbiome composition and diversity included maternal antibiotic and probiotic uses, dietary intake, pre-pregnancy body mass index (BMI), gestational weight gain (GWG), diabetes, mood, and others. Eleven studies contributed to three meta-analyses quantifying associations between maternal intrapartum antibiotic exposure (IAP), BMI and GWG, and infant microbiome alpha diversity (Shannon Index). IAP, maternal overweight/obesity and excessive GWG were all associated with reduced diversity. Most studies were observational, few included early recruitment or longitudinal follow-up, and the timing, frequency, and methodologies related to stool sampling and analysis were variable. Standardization and collaboration are imperative to enhance understanding in this complex and rapidly evolving area.
Collapse
Affiliation(s)
- Allison Grech
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Clare E Collins
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW, Australia
| | - Andrew Holmes
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Camperdown, NSW, Australia
| | - Ravin Lal
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Kerith Duncanson
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Rachael Taylor
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW, Australia
| | - Adrienne Gordon
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
29
|
Łubiech K, Twarużek M. Lactobacillus Bacteria in Breast Milk. Nutrients 2020; 12:E3783. [PMID: 33321792 PMCID: PMC7764098 DOI: 10.3390/nu12123783] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Breast milk is an optimal food for infants and toddlers. The composition of breast milk adapts to the needs of the developing organism, satisfying nutritional needs at an early stage of growth and development. The results of research to date have shown that breast milk is the best food for a child, containing not only nutrients but also biologically active substances that aid in the optimal, proper growth and development of infants. Among the many components of breast milk, an important element is the probiotic microflora, including bacteria of the genus Lactobacillus spp. These organisms exert a multidirectional, health-promoting effect on the body of children who consume breast milk. The number of lactic acid bacteria, including Lactobacillus, colonizing the breast milk environment and their species diversity varies and depends on many factors, both maternal and environmental. Breast milk, as a recommended food for infants, is an important source of probiotic microflora. The aim of this study was to present the current understanding of probiotic bacteria of the genus Lactobacillus present in breast milk.
Collapse
Affiliation(s)
- Katarzyna Łubiech
- Department of Physiology and Toxicology, Faculty of Biological Sciences, Kazimierz Wielki University, Chodkiewicza 30 St., 85-064 Bydgoszcz, Poland;
| | | |
Collapse
|
30
|
Fernández L, Pannaraj PS, Rautava S, Rodríguez JM. The Microbiota of the Human Mammary Ecosystem. Front Cell Infect Microbiol 2020; 10:586667. [PMID: 33330129 PMCID: PMC7718026 DOI: 10.3389/fcimb.2020.586667] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk contains a dynamic and complex site-specific microbiome, which is not assembled in an aleatory way, formed by organized microbial consortia and networks. Presence of some genera, such as Staphylococcus, Streptococcus, Corynebacterium, Cutibacterium (formerly known as Propionibacterium), Lactobacillus, Lactococcus and Bifidobacterium, has been detected by both culture-dependent and culture-independent approaches. DNA from some gut-associated strict anaerobes has also been repeatedly found and some studies have revealed the presence of cells and/or nucleic acids from viruses, archaea, fungi and protozoa in human milk. Colostrum and milk microbes are transmitted to the infant and, therefore, they are among the first colonizers of the human gut. Still, the significance of human milk microbes in infant gut colonization remains an open question. Clinical studies trying to elucidate the question are confounded by the profound impact of non-microbial human milk components to intestinal microecology. Modifications in the microbiota of human milk may have biological consequences for infant colonization, metabolism, immune and neuroendocrine development, and for mammary health. However, the factors driving differences in the composition of the human milk microbiome remain poorly known. In addition to colostrum and milk, breast tissue in lactating and non-lactating women may also contain a microbiota, with implications in the pathogenesis of breast cancer and in some of the adverse outcomes associated with breast implants. This and other open issues, such as the origin of the human milk microbiome, and the current limitations and future prospects are addressed in this review.
Collapse
Affiliation(s)
- Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Pia S. Pannaraj
- Department of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine and Children’s Hospital, Los Angeles, CA, United States
| | - Samuli Rautava
- University of Helsinki and Helsinki University Hospital, New Children’s Hospital, Pediatric Research Center, Helsinki, Finland
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
31
|
Moossavi S, Azad MB. Origins of human milk microbiota: new evidence and arising questions. Gut Microbes 2020; 12:1667722. [PMID: 31684806 PMCID: PMC7524145 DOI: 10.1080/19490976.2019.1667722] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/22/2019] [Accepted: 09/10/2019] [Indexed: 02/08/2023] Open
Abstract
Human milk contains a diverse community of bacteria. The growing appreciation of commensal microbes and increasing availability of high-throughput technology has set the stage for a theory-driven approach to the study of milk microbiota, and translation of this knowledge to improve maternal and child health. We recently profiled the milk microbiota of healthy Canadian mothers and applied theory-driven causal modeling, finding that mode of breast milk feeding (nursing directly at the breast vs. pumping and feeding breast milk from a bottle) was significantly associated with milk microbiota composition. This observation could reflect an increased exposure to pumps and/or a decreased exposure to the infant mouth. Either way, it provides evidence for the retrograde mechanism of milk inoculation. Here, we discuss the implications of this research and related controversies, and raise new questions about the origins and function of milk bacteria.
Collapse
Affiliation(s)
- Shirin Moossavi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Winnipeg, MB, Canada
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Meghan B. Azad
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Winnipeg, MB, Canada
- Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
32
|
The Impact of the Postpartum "Doing-the-Month" Practice on Human Milk Microbiota: A Pilot Study in Taiwan. Microorganisms 2020; 8:microorganisms8091283. [PMID: 32842688 PMCID: PMC7564682 DOI: 10.3390/microorganisms8091283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/26/2022] Open
Abstract
Human milk microorganisms could benefit the healthy development of the immune system in infants. In Asia, the practice of “doing-the-month” indicates a month-long period of postpartum recuperation for new mothers. This is composed of cultural practices, traditional beliefs, behavioral, dietary, and herbal therapies. In this pilot study, we evaluated the effect of “doing-the-month” on the human milk microbiota using a molecular approach. We collected two “doing-the-month” milk groups from randomly recruited mothers who had completed their “doing-the-month” program in either postpartum care center A (milk-PCA, n = 14) or postpartum care center B (milk-PCB, n = 27) for 20 to 30 days. As for the control group, milk samples were selected from postpartum mothers (milk-H, n = 46), who did not conduct the “doing-the-month” program. We found that the “doing-the-month” milk samples were associated with more diverse and unique milk microbiota and that these samples were also linked with more abundant Lactobacillus (milk-PCB) and prevalent Bifidobacteria (milk-PCA and milk-PCB). In addition, the milk samples from “doing-the-month” mothers could be enriched with more Archaea bacterial members, but the “non-doing-the-month” milk samples were enriched with more common skin-, oral-, and environmental-related bacterial members. This study highlights the impact maternal practices may have on the milk microbiome. More research is needed to investigate the effects this may have on infant immune health.
Collapse
|
33
|
Effect of Sample Collection (Manual Expression vs. Pumping) and Skimming on the Microbial Profile of Human Milk Using Culture Techniques and Metataxonomic Analysis. Microorganisms 2020; 8:microorganisms8091278. [PMID: 32825795 PMCID: PMC7564974 DOI: 10.3390/microorganisms8091278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
Human milk microbiota is a unique bacterial community playing a relevant role in infant health, but its composition depends on different factors (woman health, lactation stage, and geographical lactation). However, information is lacking regarding some other factors that may affect the bacterial community of human milk. In this study we aimed to study the impact of the sample collection method and the skimming procedure using culture-dependent and culture-independent techniques to study the human milk microbial profile. One set of milk samples was provided by women (n = 10) in two consecutive days; half of the samples were collected the first day by manual expression and the other half on the second day by pumping. The rest of the participants (n = 17) provided milk samples that were fractionated by centrifugation; the bacterial profiles of whole milk and skimmed milk were compared by culture techniques in 10 milk samples, while those of whole milk, fat and skimmed milk were subjected to metataxonomic analysis in seven samples. Globally, the results obtained revealed high interindividual variability but that neither the use of single-use sterile devices to collect the sample nor the skimming procedure have a significant impact of the microbial profile of human samples.
Collapse
|
34
|
Ding J, Liao N, Zheng Y, Yang L, Zhou H, Xu K, Han C, Luo H, Qin C, Tang C, Wei L, Meng H. The Composition and Function of Pigeon Milk Microbiota Transmitted From Parent Pigeons to Squabs. Front Microbiol 2020; 11:1789. [PMID: 32849405 PMCID: PMC7417789 DOI: 10.3389/fmicb.2020.01789] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Mammalian neonates obtain antibodies, nutrients, and microbiota from breast milk that help them resist the complex growth environment. Similar to mammals' lactation behavior for their offspring, parent pigeons regurgitate pigeon milk (PM) from their crops to feed the squabs. Whether pigeon milk is as valuable as mammalian milk is not clear, especially in terms of microbiota. This study adopted 16S rRNA gene sequencing to investigate the microbial composition and function in pigeon milk. We found abundant microbiota in pigeon milk. The dominant genera in parent pigeons' milk were Lactobacillus, Enterococcus, Veillonella, and Bifidobacterium. An analysis of squab milk (SM) showed that Lactobacillus also accounted for a considerable proportion, followed by Bifidobacterium. Most of the squab milk microbial genera were also detected in parent pigeons. Microbial functional analysis showed that the squab milk microbes were more involved in the pathways of carbohydrate metabolism, amino acid metabolism, and energy metabolism. These findings indicated that microbiota play an important role in squabs and can be transmitted from parent pigeons to squabs by pigeon milk. The presence of plentiful probiotics in squabs also suggests that adding probiotics in artificial pigeon milk may promote the growth and development of squabs and improve the production performance of pigeons.
Collapse
Affiliation(s)
- Jinmei Ding
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Liao
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuming Zheng
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Lingyu Yang
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Zhou
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Xu
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chengxiao Han
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Huaixi Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chao Qin
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chunhong Tang
- Shanghai Xinrong Big Emperor Pigeon Breeding Professional Cooperation, Shanghai, China
| | - Longxing Wei
- Fengxian District Animal Disease Prevention and Control Center, Shanghai, China
| | - He Meng
- Shanghai Key Laboratory of Veterinary Biotechnology, Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
O'Neill IJ, Sanchez Gallardo R, Saldova R, Murphy EF, Cotter PD, McAuliffe FM, van Sinderen D. Maternal and infant factors that shape neonatal gut colonization by bacteria. Expert Rev Gastroenterol Hepatol 2020; 14:651-664. [PMID: 32552141 DOI: 10.1080/17474124.2020.1784725] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Early life is a critical developmental window coinciding with the establishment of a community of neonatal gut microbes which are vitally important for immune development. The composition of this microbial community is affected by multiple factors. AREAS COVERED The effect of pre-pregnancy and pregnancy maternal health, maternal nutrition, pregnancy disorders such as gestational diabetes, maternal antibiotic usage, delivery mode, infant feeding, and infant antibiotic usage on gut microbial composition are outlined along with the potential impact of associated microbiota differences on infant health. EXPERT OPINION Recent developments in understanding what shapes our microbiota indicates that the greatest impact on infant gut microbiota composition during the first year of life is seen with the mode of delivery, infant diet, and infant antibiotic usage. Current data is insufficient to fully establish the role of apparently less important factors such as maternal health on microbiota development although their impact is likely smaller. Technological advances will allow for improved understanding of underlying mechanisms by which specific microbes impact on infant health, which in time will enable full appreciation of the role of the gut microbiota in early life development.
Collapse
Affiliation(s)
- Ian J O'Neill
- APC Microbiome Ireland, National University of Ireland , Cork, Ireland
| | - Rocio Sanchez Gallardo
- APC Microbiome Ireland, National University of Ireland , Cork, Ireland.,School of Microbiology, National University of Ireland , Cork, Ireland
| | - Radka Saldova
- The National Institute for Bioprocessing, Research, and Training (NIBRT) , Dublin, Ireland.,UCD School of Medicine, College of Health and Agricultural Science, University College Dublin , Dublin, Ireland
| | - Eileen F Murphy
- Alimentary Health Group, Cork Airport Business Park , Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, National University of Ireland , Cork, Ireland.,Teagasc Food Research Centre , Cork, Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital , Dublin, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, National University of Ireland , Cork, Ireland.,School of Microbiology, National University of Ireland , Cork, Ireland
| |
Collapse
|
36
|
Navarro-Tapia E, Sebastiani G, Sailer S, Toledano LA, Serra-Delgado M, García-Algar Ó, Andreu-Fernández V. Probiotic Supplementation During the Perinatal and Infant Period: Effects on Gut Dysbiosis and Disease. Nutrients 2020; 12:E2243. [PMID: 32727119 PMCID: PMC7468726 DOI: 10.3390/nu12082243] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The perinatal period is crucial to the establishment of lifelong gut microbiota. The abundance and composition of microbiota can be altered by several factors such as preterm delivery, formula feeding, infections, antibiotic treatment, and lifestyle during pregnancy. Gut dysbiosis affects the development of innate and adaptive immune responses and resistance to pathogens, promoting atopic diseases, food sensitization, and infections such as necrotizing enterocolitis (NEC). Recent studies have indicated that the gut microbiota imbalance can be restored after a single or multi-strain probiotic supplementation, especially mixtures of Lactobacillus and Bifidobacterium strains. Following the systematic search methodology, the current review addresses the importance of probiotics as a preventive or therapeutic tool for dysbiosis produced during the perinatal and infant period. We also discuss the safety of the use of probiotics in pregnant women, preterm neonates, or infants for the treatment of atopic diseases and infections.
Collapse
Affiliation(s)
- Elisabet Navarro-Tapia
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
| | - Giorgia Sebastiani
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Sebastian Sailer
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Laura Almeida Toledano
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, 08950 Barcelona, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Valencian International University (VIU), 46002 Valencia, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, 08028 Barcelona, Spain
| |
Collapse
|
37
|
Effects of probiotics on salivary cytokines and immunoglobulines: a systematic review and meta-analysis on clinical trials. Sci Rep 2020; 10:11800. [PMID: 32678117 PMCID: PMC7366729 DOI: 10.1038/s41598-020-67037-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/02/2020] [Indexed: 11/09/2022] Open
Abstract
Findings on the effects of probiotics on salivary cytokines and immunoglobulines have been conflicting. We aimed to perform a systematic review and meta-analysis on clinical trials that examined the effects of oral intake and local administration of probiotics on salivary cytokines and immunoglobulines in adults. We searched PubMed, MEDLINE, SCOPUS, EMBASE, and Google Scholar up to April 2020 for all relevant published papers assessing probiotic intakes and salivary cytokines and immunoglobulines. We included all randomized clinical trials that investigated the effect of oral probiotic supplementation or lozenges tablets on inflammatory biomarkers in adults. Studies that reported their effect sizes as mean ± SD or mean ± SEM were included. After excluding non-relevant papers, 8 studies remained in this review. Combining findings from 3 studies with 4 effect sizes, we found no significant reduction in salivary IgA concentrations after oral probiotic supplementation [weighted mean difference (WMD): -0.26; 95% CI: (-0.86, 0.35)]. A significant increase in salivary IL-1β concentrations reached after local probiotic supplementation (WMD: 28.21; 95% CI: 18.42, 38.01); however, no significant changes in salivary IL-6 concentrations after local probiotic supplementation was found (WMD: 0.36; 95% CI: -0.85, 1.56). We observed a significant increase in salivary IL-8 concentrations after local probiotic supplementation (WMD: 31.82; 95% CI: 27.56, 36.08). In case of salivary IL-10 concentrations after local probiotic administration, no significant reduction was seen (WMD: -0.02; 95% CI: -0.10, 0.06). we found that oral and local administrations of probiotics might influence some of salivary cytokines. However, additional clinical trials are required to examine these effects on further pro- and anti-inflammatory cytokines and immunoglobulines.
Collapse
|
38
|
Kapourchali FR, Cresci GAM. Early-Life Gut Microbiome-The Importance of Maternal and Infant Factors in Its Establishment. Nutr Clin Pract 2020; 35:386-405. [PMID: 32329544 DOI: 10.1002/ncp.10490] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
The early-life microbiome is gaining appreciation as a major influencer in human development and long-term health. Multiple factors are known to influence the initial colonization, development, and function of the neonatal gut microbiome. In addition, alterations in early-life gut microbial composition is associated with several chronic health conditions such as obesity, asthma, and allergies. In this review, we focus on both maternal and infant factors known to influence early-life gut colonization. Also reviewed is the important role of infant feeding, including evidence-based strategies for maternal and infant supplementation with the goal to protect and/or restore the infant gut microbiome.
Collapse
Affiliation(s)
| | - Gail A M Cresci
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Human Nutrition, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
39
|
Dzidic M, Mira A, Artacho A, Abrahamsson TR, Jenmalm MC, Collado MC. Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life. Pediatr Allergy Immunol 2020; 31:250-257. [PMID: 31736150 DOI: 10.1111/pai.13176] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/27/2022]
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.
Collapse
Affiliation(s)
- Majda Dzidic
- Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA-CSIC), Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain.,Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain.,CIBER-ESP, Madrid, Spain
| | - Alejandro Artacho
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO, Valencia, Spain
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine and Department of Pediatrics, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology (IATA-CSIC), Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| |
Collapse
|
40
|
Azagra-Boronat I, Tres A, Massot-Cladera M, Franch À, Castell M, Guardiola F, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Lactobacillus fermentum CECT5716 supplementation in rats during pregnancy and lactation affects mammary milk composition. J Dairy Sci 2020; 103:2982-2992. [PMID: 32008776 DOI: 10.3168/jds.2019-17384] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022]
Abstract
Lactobacillus fermentum CECT5716 has shown immunomodulatory action and reduction of infections; therefore, it is suggested to be appropriate for use in early life. The present study aimed to assess the effects of the supplementation of L. fermentum CECT5716 in rats during gestation and lactation periods on the composition of some mammary milk components such as microbiota, fatty acid (FA) profile, and immunoglobulins. Wistar rats were supplemented by oral gavage with 1010 cfu/d of Lactobacillus fermentum CECT5716 (n = 6) or vehicle (n = 6) for 5 wk, comprising the 3 wk of gestation and the first 2 wk of lactation. At the end of the intervention, milk, mammary glands, and cecal contents were obtained for the tracking of the probiotic strain by nested PCR-quantitative PCR. Additionally, milk samples were used for the analysis of microbiota by 16S rRNA sequencing, FA by gas chromatography-flame ionization detector, and immunoglobulin by Luminex (Luminex Corporation, Austin, TX). Although L. fermentum CECT5716 administration did not modify the overall composition of milk microbiota, the strain was detected in 50% of the milk samples of rats supplemented with the probiotic. Moreover, probiotic administration induced beneficial changes in the FA composition of milk by increasing total PUFA, including linoleic and α-linolenic acids, and decreasing the proportion of palmitic acid. Finally, the milk of the rats treated with the probiotic showed a 2-fold increase of IgA levels. The supplementation with L. fermentum CECT5716 during pregnancy and lactation periods improved the milk composition of FA and immunoglobulins. These effects were not linked to the presence of the strain in milk, thus suggesting that the mechanism is connected to intestinal compartment. These findings provide novel insight into a potential new approach for infants to benefit from better nutrition, development of a healthy immune system and microbiota, and protection from gastrointestinal infections.
Collapse
Affiliation(s)
- Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain
| | - Alba Tres
- Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain; Department of Nutrition, Food Science and Gastronomy, Torribera Food Science Campus, Faculty of Pharmacy and Food Science, University of Barcelona, Santa Coloma de Gramenet 08921, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain
| | - Francesc Guardiola
- Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain; Department of Nutrition, Food Science and Gastronomy, Torribera Food Science Campus, Faculty of Pharmacy and Food Science, University of Barcelona, Santa Coloma de Gramenet 08921, Spain
| | - Francisco J Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain.
| | - M José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona 08028, Spain; Nutrition and Food Safety Research Institute, Santa Coloma de Gramenet 08921, Spain
| |
Collapse
|
41
|
Lee YJ, Kim KM, Jung HL, Shim JY, Kim DS, Shim JW. Relationship between Breastfeeding, Birth History, and Acute Pyelonephritis in Infants. J Korean Med Sci 2020; 35:e32. [PMID: 32103642 PMCID: PMC7049624 DOI: 10.3346/jkms.2020.35.e32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/11/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Although the clinical importance of the immunological benefits of breastfeeding has been emphasized for decades, their direct relationship with acute pyelonephritis (APN) is still not clear. Our goal was to determine whether breastfeeding truly provides protection against APNs, while investigating the effects of other factors such as sex, age, mode of delivery, and birth weight on APN. METHODS A total of 62 infants under 6 months of age who had both microbiologically and radiologically-confirmed APN were enrolled in the case group. Healthy infants (n = 178) who visited the hospital for scheduled vaccinations were enrolled in the control group. The following participant characteristics were compared between the case and control groups: age, sex, birth order among siblings, feeding methods, weight percentile by month, birth weight percentile by gestational age, gestational age at birth, and mode of delivery. RESULTS Babies exclusively fed with manufactured infant formulae before 6 months of age had significantly higher risk for APN than breastfed or mixed-fed infants (odds ratio [OR], 3.4; 95% confidence interval [CI], 1.687-7.031; P = 0.001). Firstborn babies had lower risk for APN than 2nd- or 3rd-born babies (OR, 0.43; 95% CI, 0.210-0.919). Other factors that increased the risk for APN were low birth weight percentiles (OR, 8.33; 95% CI, 2.300-30.166) and birth via caesarean section (OR, 2.32; 95% CI, 1.097-4.887). There were more preterm births in the case group (10.9% vs. 1.7%; P = 0.002), but this did not increase the risk for APN (OR, 4.47; P = 0.063). CONCLUSION Feeding exclusively with formula before 6 months of age was related to higher risk for APN, which demonstrates that breastfeeding has a protective effect against APN. The other risk factors for APN were birth order (≥ 2nd-born), low birth weight, and birth via caesarean section.
Collapse
Affiliation(s)
- Young Ju Lee
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Moon Kim
- Department of Pediatrics, Hwacheon Health Center and County Hospital, Hwacheon, Korea
| | - Hye Lim Jung
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Yeon Shim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Deok Soo Kim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Won Shim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
42
|
Liu W, Chen M, Duo L, Wang J, Guo S, Sun H, Menghe B, Zhang H. Characterization of potentially probiotic lactic acid bacteria and bifidobacteria isolated from human colostrum. J Dairy Sci 2020; 103:4013-4025. [PMID: 32113772 DOI: 10.3168/jds.2019-17602] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/26/2019] [Indexed: 01/02/2023]
Abstract
Breast milk is the main source of nutrition for infants; it contains considerable microflora that can be transmitted to the infant endogenously or by breastfeeding, and it plays an important role in the maturation and development of the immune system. In this study, we isolated and identified lactic acid bacteria (LAB) from human colostrum, and screened 2 strains with probiotic potential. The LAB isolated from 40 human colostrum samples belonged to 5 genera: Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, and Staphylococcus. We also isolated Propionibacterium and Actinomyces. We identified a total of 197 strains of LAB derived from human colostrum based on their morphology and 16S rRNA sequence, among them 8 strains of Bifidobacterium and 10 strains of Lactobacillus, including 3 Bifidobacterium species and 4 Lactobacillus species. The physiological and biochemical characteristics of strains with good probiotic characteristics were evaluated. The tolerances of some of the Bifidobacterium and Lactobacillus strains to gastrointestinal fluid and bile salts were evaluated in vitro, using the probiotic strains Bifidobacterium lactis BB12 and Lactobacillus rhamnosus GG as controls. Among them, B. lactis Probio-M8 and L. rhamnosus Probio-M9 showed survival rates of 97.25 and 78.33% after digestion for 11 h in artificial gastrointestinal juice, and they exhibited growth delays of 0.95 and 1.87 h, respectively, in 0.3% bile salts. These two strains have the potential for application as probiotics and will facilitate functional studies of probiotics in breast milk and the development of human milk-derived probiotics.
Collapse
Affiliation(s)
- Wenjun Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Meixuan Chen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Lana Duo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jicheng Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Shuai Guo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Haotian Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Bilige Menghe
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
43
|
Szari S, Quinn JA. Supporting a Healthy Microbiome for the Primary Prevention of Eczema. Clin Rev Allergy Immunol 2020; 57:286-293. [PMID: 31309394 DOI: 10.1007/s12016-019-08758-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Eczema is increasing worldwide with associated increases in health costs and decreases in quality of life. There are many factors that are speculated to interact in the development of eczema including genetics and environmental exposures. Prevention of the development of eczema may prevent the further development of food allergies and asthma. This concept has prompted a variety of research into the area of primary prevention of eczema in infants. This exploration includes a growing body of research examining infants supplemented with probiotics, prebiotics, or both (synbiotics) often compared with their breastfed counterparts. The goal of this paper is to examine the evidence for manipulating the microbiome in the prevention of eczema. Several strains of probiotics, compositions of prebiotics, and varied combinations of both are commercially available. Evidence supports altering the microbiome in infants at high risk of atopy who are not able to breastfeed with Lactobacillus strains when given both prenatally followed by prolonged use (greater than 6 months) postnatally for the primary prevention of eczema. Prebiotics have also been shown beneficial for primary prevention of eczema in formula-fed infants with prolonged use greater than 6 months. These findings are in keeping with the World Allergy Organization (WAO) recommendations that support interventions to manipulate the microbiome with both probiotics and prebiotics.
Collapse
Affiliation(s)
- Sofia Szari
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA.
| | - James A Quinn
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA
| |
Collapse
|
44
|
Kao HF, Wang YC, Tseng HY, Wu LSH, Tsai HJ, Hsieh MH, Chen PC, Kuo WS, Liu LF, Liu ZG, Wang JY. Goat Milk Consumption Enhances Innate and Adaptive Immunities and Alleviates Allergen-Induced Airway Inflammation in Offspring Mice. Front Immunol 2020; 11:184. [PMID: 32132998 PMCID: PMC7040033 DOI: 10.3389/fimmu.2020.00184] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/23/2020] [Indexed: 11/13/2022] Open
Abstract
Goat milk (GM), as compared to cow milk (CM), is easier for humans to digest. It also has antioxidant and anti-inflammatory effects and can improve minor digestive disorders and prevent allergic diseases in infants. It is unclear whether GM consumed in pregnant mothers has any protective effects on allergic diseases in infants. In this experimental study with mice, we found GM feeding enhanced immunoglobulin production, antigen-specific (ovalbumin, OVA) immune responses, and phagocytosis activity. The GM-fed mice had an increasing proportion of CD3+ T lymphocytes in the spleen. Splenocytes isolated from these animals also showed significantly increased production of cytokines IFN-γ and IL-10. More importantly, GM feeding during pregnancy and lactation periods can confer protective activity onto offspring by alleviating the airway inflammation of allergic asthma induced by mite allergens. There was a remarkably different composition of gut microbiota between offspring of pregnant mice fed with water or with milk (GM or CM). There was a greater proportion of beneficial bacterial species, such as Akkermansia muciniphila, Bacteroides eggerthii, and Parabacteroides goldsteinii in the gut microbiota of offspring from GM- or CM-fed pregnant mice compared to the offspring of water-fed pregnant mice. These results suggested that improving the nutrition of pregnant mice can promote immunological maturation and colonization of gut microbiota in offspring. This mother-to-child biological action may provide a protective effect on atopy development and alleviate allergen-induced airway inflammation in offspring.
Collapse
Affiliation(s)
- Hui-Fang Kao
- Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan
| | - Yu-Chin Wang
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Ying Tseng
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Hui-Ju Tsai
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Miao-Hsi Hsieh
- Graduate Institute of Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chi Chen
- Graduate Institute of Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Shou Kuo
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing, China
| | - Li-Fan Liu
- Institute of Gerontology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Zhi-Gang Liu
- Department of Respirology and Allergy, Third Affiliated Hospital of Shengzhen University, Shengzhen, China
| | - Jiu-Yao Wang
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
45
|
Zimmermann P, Curtis N. Breast milk microbiota: A review of the factors that influence composition. J Infect 2020; 81:17-47. [PMID: 32035939 DOI: 10.1016/j.jinf.2020.01.023] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 01/31/2023]
Abstract
Breastfeeding is associated with considerable health benefits for infants. Aside from essential nutrients, immune cells and bioactive components, breast milk also contains a diverse range of microbes, which are important for maintaining mammary and infant health. In this review, we summarise studies that have investigated the composition of the breast milk microbiota and factors that might influence it. We identified 44 studies investigating 3105 breast milk samples from 2655 women. Several studies reported that the bacterial diversity is higher in breast milk than infant or maternal faeces. The maximum number of each bacterial taxonomic level detected per study was 58 phyla, 133 classes, 263 orders, 596 families, 590 genera, 1300 species and 3563 operational taxonomic units. Furthermore, fungal, archaeal, eukaryotic and viral DNA was also detected. The most frequently found genera were Staphylococcus, Streptococcus Lactobacillus, Pseudomonas, Bifidobacterium, Corynebacterium, Enterococcus, Acinetobacter, Rothia, Cutibacterium, Veillonella and Bacteroides. There was some evidence that gestational age, delivery mode, biological sex, parity, intrapartum antibiotics, lactation stage, diet, BMI, composition of breast milk, HIV infection, geographic location and collection/feeding method influence the composition of the breast milk microbiota. However, many studies were small and findings sometimes contradictory. Manipulating the microbiota by adding probiotics to breast milk or artificial milk offers an exciting avenue for future interventions to improve infant health.
Collapse
Affiliation(s)
- Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia.
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| |
Collapse
|
46
|
Kordy K, Gaufin T, Mwangi M, Li F, Cerini C, Lee DJ, Adisetiyo H, Woodward C, Pannaraj PS, Tobin NH, Aldrovandi GM. Contributions to human breast milk microbiome and enteromammary transfer of Bifidobacterium breve. PLoS One 2020; 15:e0219633. [PMID: 31990909 PMCID: PMC6986747 DOI: 10.1371/journal.pone.0219633] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/11/2019] [Indexed: 01/08/2023] Open
Abstract
Increasing evidence supports the importance of the breast milk microbiome in seeding the infant gut. However, the origin of bacteria in milk and the process of milk microbe-mediated seeding of infant intestine need further elucidation. Presumed sources of bacteria in milk include locations of mother-infant and mother-environment interactions. We investigate the role of mother-infant interaction on breast milk microbes. Shotgun metagenomics and 16S rRNA gene sequencing identified milk microbes of mother-infant pairs in breastfed infants and in infants that have never latched. Although breast milk has low overall biomass, milk microbes play an important role in seeding the infant gut. Breast milk bacteria were largely comprised of Staphylococcus, Streptococcus, Acinetobacter, and Enterobacter primarily derived from maternal areolar skin and infant oral sites in breastfeeding pairs. This suggests that the process of breastfeeding is a potentially important mechanism for propagation of breast milk microbes through retrograde flux via infant oral and areolar skin contact. In one infant delivered via Caesarian section, a distinct strain of Bifidobacteria breve was identified in maternal rectum, breast milk and the infant’s stool potentially suggesting direct transmission. This may support the existence of microbial translocation of this anaerobic bacteria via the enteromammary pathway in humans, where maternal bacteria translocate across the maternal gut and are transferred to the mammary glands. Modulating sources of human milk microbiome seeding potentially imply opportunities to ultimately influence the development of the infant microbiome and health.
Collapse
Affiliation(s)
- Kattayoun Kordy
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
- Department of Pediatrics, University of Southern California, Los Angeles, CA, United States of America
| | - Thaidra Gaufin
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Martin Mwangi
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Fan Li
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
- Department of Pediatrics, University of Southern California, Los Angeles, CA, United States of America
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Chiara Cerini
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
| | - David J. Lee
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
| | - Helty Adisetiyo
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
| | - Cora Woodward
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Pia S. Pannaraj
- Department of Pediatrics, Children’s Hospital of Los Angeles, Los Angeles, CA, United States of America
- Department of Pediatrics, University of Southern California, Los Angeles, CA, United States of America
| | - Nicole H. Tobin
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Grace M. Aldrovandi
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
47
|
Nyangahu DD, Jaspan HB. Influence of maternal microbiota during pregnancy on infant immunity. Clin Exp Immunol 2019; 198:47-56. [PMID: 31121057 PMCID: PMC6718277 DOI: 10.1111/cei.13331] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2019] [Indexed: 12/11/2022] Open
Abstract
Microbiota from various maternal sites, including the gut, vagina and breast milk, are known to influence colonization in infants. However, emerging evidence suggests that these sites may exert their influence prior to delivery, in turn influencing fetal immune development. The dogma of a sterile womb continues to be challenged. Regardless, there is convincing evidence that the composition of the maternal gut prior to delivery influences neonatal immunity. Therefore, while the presence and function of placental microbiome is not clear, there is consensus that the gut microbiota during pregnancy is a critical determinant of offspring health. Data supporting the notion of bacterial translocation from the maternal gut to extra-intestinal sites during pregnancy are emerging, and potentially explain the presence of bacteria in breast milk. Much evidence suggests that the maternal gut microbiota during pregnancy potentially determines the development of atopy and autoimmune phenotypes in offspring. Here, we highlight the role of the maternal microbiota prior to delivery on infant immunity and predisposition to diseases. Moreover, we discuss potential mechanisms that underlie this phenomenon.
Collapse
Affiliation(s)
- D. D. Nyangahu
- Department of PediatricsUniversity of Washington and Seattle Children’s Research InstituteSeattleWAUSA
| | - H. B. Jaspan
- Department of PediatricsUniversity of Washington and Seattle Children’s Research InstituteSeattleWAUSA
- Department of Global HealthUniversity of WashingtonSeattleWAUSA
- Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, Division of ImmunologyUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
48
|
Boix-Amorós A, Collado MC, Van't Land B, Calvert A, Le Doare K, Garssen J, Hanna H, Khaleva E, Peroni DG, Geddes DT, Kozyrskyj AL, Warner JO, Munblit D. Reviewing the evidence on breast milk composition and immunological outcomes. Nutr Rev 2019; 77:541-556. [PMID: 31111150 DOI: 10.1093/nutrit/nuz019] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A large number of biologically active components have been found in human milk (HM), and in both human and animal models, studies have provided some evidence suggesting that HM composition can be altered by maternal exposures, subsequently influencing health outcomes for the breastfed child. Evidence varies from the research studies on whether breastfeeding protects the offspring from noncommunicable diseases, including those associated with immunological dysfunction. It has been hypothesized that the conflicting evidence results from HM composition variations, which contain many immune active molecules, oligosaccharides, lactoferrin, and lysozyme in differing concentrations, along with a diverse microbiome. Determining the components that influence infant health outcomes in terms of both short- and long-term sequelae is complicated by a lack of understanding of the environmental factors that modify HM constituents and thereby offspring outcomes. Variations in HM immune and microbial composition (and the differing infantile responses) may in part explain the controversies that are evidenced in studies that aim to evaluate the prevalence of allergy by prolonged and exclusive breastfeeding. HM is a "mixture" of immune active factors, oligosaccharides, and microbes, which all may influence early immunological outcomes. This comprehensive review provides an in-depth overview of existing evidence on the studied relationships between maternal exposures, HM composition, vaccine responses, and immunological outcomes.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Belinda Van't Land
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Anna Calvert
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
- Imperial College London, London, United Kingdom
- Public Health England, Porton Down, United Kingdom, and the MRC Unit, Fajara, Gambia
| | - Johan Garssen
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Ekaterina Khaleva
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- University of Southampton, Southampton, UK
| | - Diego G Peroni
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Donna T Geddes
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- School of Molecular Sciences, the University of Western Australia, Perth, Australia
| | - Anita L Kozyrskyj
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Pediatrics, Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, School of Public Health, University of Alberta, Alberta, Canada
| | - John O Warner
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- National Institute for Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, London, United Kingdom
| | - Daniel Munblit
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- Department of Pediatrics, Sechenov University, Moscow, Russia, and the Solov'ev Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
49
|
Ruiz L, García-Carral C, Rodriguez JM. Unfolding the Human Milk Microbiome Landscape in the Omics Era. Front Microbiol 2019; 10:1378. [PMID: 31293535 PMCID: PMC6604669 DOI: 10.3389/fmicb.2019.01378] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 06/03/2019] [Indexed: 12/31/2022] Open
Abstract
Studies conducted in the last years have demonstrated that human milk represents a continuous supply of beneficial bacteria to the infant gut, which contribute to the maturation of the digestive and immune functions in the developing infant. Nevertheless, the origin of bacterial populations in milk is not fully understood yet and they have been proposed to originate from maternal skin, infant’s mouth, and (or) endogenously, from the maternal digestive tract through a mechanism involving immune cells. Understanding the composition, functions and assembly of the human milk microbiota has important implications not only for the infant gut microbiota establishment, but also for the mammary health since dysbiosis in the milk bacteria may lead to mastitis. Besides, host, microbial, medical and environmental factors may affect the composition of the human milk microbiome, with implications for the mother-infant health. Application of both culture-dependent and -independent techniques to assess the milk microbiome faces some practical limitations but, together, have allowed providing novel and complementary views on its origin, composition and functioning as summarized in this minireview. In the next future, the application of the ultimate advances in next-generation sequencing and omics approaches, including culturomics, will allow a detailed and comprehensive understanding of the composition and functions of these microbial communities, including their interactions with other milk components, expanding the opportunities to design novel microbiome-based modulation strategies for this ecosystem.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
50
|
Ruiz L, Bacigalupe R, García-Carral C, Boix-Amoros A, Argüello H, Silva CB, de Los Angeles Checa M, Mira A, Rodríguez JM. Microbiota of human precolostrum and its potential role as a source of bacteria to the infant mouth. Sci Rep 2019; 9:8435. [PMID: 31182726 PMCID: PMC6557856 DOI: 10.1038/s41598-019-42514-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Human milk represents a source of bacteria for the initial establishment of the oral (and gut) microbiomes in the breastfed infant, however, the origin of bacteria in human milk remains largely unknown. While some evidence points towards a possible endogenous enteromammary route, other authors have suggested that bacteria in human milk are contaminants from the skin or the breastfed infant mouth. In this work 16S rRNA sequencing and bacterial culturing and isolation was performed to analyze the microbiota on maternal precolostrum samples, collected from pregnant women before delivery, and on oral samples collected from the corresponding infants. The structure of both ecosystems demonstrated a high proportion of taxa consistently shared among ecosystems, Streptococcus spp. and Staphylococcus spp. being the most abundant. Whole genome sequencing on those isolates that, belonging to the same species, were isolated from both the maternal and infant samples in the same mother-infant pair, evidenced that in 8 out of 10 pairs both isolates were >99.9% identical at nucleotide level. The presence of typical oral bacteria in precolostrum before contact with the newborn indicates that they are not a contamination from the infant, and suggests that at least some oral bacteria reach the infant's mouth through breastfeeding.
Collapse
Affiliation(s)
- Lorena Ruiz
- IPLA-CSIC, Department of Microbiology and Biochemistry of Dairy Products, Institute of Dairy Products of Asturias, Villaviciosa, Spain. .,Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.
| | - Rodrigo Bacigalupe
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.,Probisearch S.L., C/Santiago Grisolía, 2, 28760, Tres Cantos, Spain
| | - Alba Boix-Amoros
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain
| | - Héctor Argüello
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Camilla Beatriz Silva
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.,Universidade de Uberaba, Uberaba, Brazil
| | | | - Alex Mira
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain.
| | - Juan M Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.
| |
Collapse
|