1
|
Tas GG, Sati L. Probiotic Lactobacillus rhamnosus species: considerations for female reproduction and offspring health. J Assist Reprod Genet 2024; 41:2585-2605. [PMID: 39172320 PMCID: PMC11535107 DOI: 10.1007/s10815-024-03230-6] [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: 06/26/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
Lactobacillus rhamnosus is a type of bacteria known as a probiotic and is often used to support the health of the digestive system and vaginal flora. This type of bacteria has an important role, showing positive effects on female reproductive biology, particularly by maintaining the balance of microorganisms in the vagina, reducing the risk of infection, and strengthening the immune system to support maternal health during pregnancy. There are also studies showing that these probiotics prevent maternal obesity and gestational diabetes. Consuming probiotics containing Lactobacillus rhamnosus strains may support the intestinal health of breastfeeding mothers, but they may also contribute to the health of offspring. Therefore, this review focuses on the current available data for examining the effects of Lactobacillus rhamnosus strains on female reproductive biology and offspring health. A systematic search was conducted in the PubMed and Web of Science databases from inception to May 2024. The search strategy was performed using keywords and MeSH (Medical Subject Headings) terms. Inconsistent ratings were resolved through discussion. This review is strengthened by multiple aspects of the methodological approach. The systematic search strategy, conducted by two independent reviewers, enabled the identification and evaluation of all relevant literature. Although there is a limited number of studies with high heterogeneity, current literature highlights the important contribution of Lactobacillus rhamnosus probiotics in enhancing female reproductive health and fertility. Furthermore, the probiotic bacteria in breast milk may also support the intestinal health of newborn, strengthen the immune system, and protect them against diseases at later ages.
Collapse
Affiliation(s)
- Gizem Gamze Tas
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Leyla Sati
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
2
|
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
|
3
|
Snaidr L, Mühlhahn P, Beimfohr C, Kreuzer C, Richly C, Snaidr J. Specific cultivation-independent enumeration of viable cells in probiotic products using a combination of fluorescence in situ hybridization and flow cytometry. Front Microbiol 2024; 15:1410709. [PMID: 38933029 PMCID: PMC11199854 DOI: 10.3389/fmicb.2024.1410709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
This study introduces an optimized integration of flow cytometry and fluorescence in situ hybridization (Flow-FISH) as an approach for the specific enumeration of gram-positive bacteria in probiotic products, overcoming the limitations of conventional methods. The enhanced Flow-FISH technique synergizes the rapid and automated capabilities of flow cytometry with the high specificity of FISH, facilitating the differentiation of viable cells at the species level within probiotic blends. By analyzing lyophilized samples of Lacticaseibacillus rhamnosus, Lactiplantibacillus plantarum, and Bifidobacterium animalis subsp. lactis, and a commercial product, the study highlights the optimized Flow-FISH protocol's advantages, including reduced hybridization times to 1.5 h and elimination of centrifugation steps. Comparative evaluations with the widely accepted enumeration methods plate count and Live/Dead (L/D) staining were conducted. The study revealed that Flow-FISH produces higher viable cell counts than plate count, thereby challenging the traditional "gold standard" by highlighting its predisposition to underestimate actual viable cell numbers. Against L/D staining, Flow-FISH achieved comparable results, which, despite the different foundational premises of each technique, confirms the accuracy and reliability of our method. In conclusion, the optimized Flow-FISH protocol represents a significant leap forward in probiotic research and quality control. This method provides a rapid, robust, and highly specific alternative for the enumeration of probiotic bacteria, surpassing traditional methodologies. Its ability to enable a more detailed and reliable analysis of probiotic products paves the way for precise quality control and research insights, underscoring its potential to improve the field significantly.
Collapse
|
4
|
Li Z, Zhou Q, Qingsong Q, Liao Y, Yang F, Sheng M, Feng L, Shi X. Effect of Maifan Stone on the Growth of Probiotics and Regulation of Gut Microbiota. Lett Appl Microbiol 2022; 75:1423-1432. [PMID: 35975465 DOI: 10.1111/lam.13809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/02/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022]
Abstract
Maifan stone is a kind of mineral medicine in Chinese medicine, which has good adsorption, dissolution, mineralization, and biological activity. It has an excellent therapeutic effect on livestock, poultry, and aquatic animals suffering from intestinal diseases. This study explored the effect of Maifan stone on the growth ability of Lacticaseibacillus rhamnosus GG (L. rhamnosus GG) and the effect of Maifan stone-L. rhamnosus GG fermented product on the intestinal inflammation and gut microbiota. We find that Maifan stone can adsorb L. rhamnosus GG to form a carrier bacteria. Maifan stone has the characteristics of acid tolerance and bile salt tolerance and can also improve the activity of L. rhamnosus GG in artificial gastrointestinal juice. The fermented product can reduce the degree of diarrhea and colon pathology in rats to a certain extent and significantly improve intestinal inflammatory factors and gut microbiota. This study improves the application effect of L. rhamnosus GG in the prevention and treatment of diarrhea animals and provide a scientific basis for the rational development of Maifan stone resources.
Collapse
Affiliation(s)
- ZhiXun Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qing Zhou
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qu Qingsong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yuyao Liao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Fang Yang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mengke Sheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Lei Feng
- Inner Mongolia Yougaoya Health Technology Co., Ltd., Inner Mongolia, 028399, China
| | - Xinyuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.,Key Laboratory for Production Process Control and Quality Evaluation of Traditional Chinese Medicine, Beijing Municipal Science & Technology Commission, Beijing, 100029, China
| |
Collapse
|
5
|
Wang D, Zhang T, Hao H, Zhang H, Ye H, Zhao C. Probiotic properties of a Spaceflight-induced mutant Lactobacillus plantarum SS18-50 in mice. Endocr Metab Immune Disord Drug Targets 2021; 22:525-531. [PMID: 34533451 DOI: 10.2174/1871530321666210917163719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Probiotics are a group of bacteria that play a critical role in intestinal microbiota homeostasis and may help adjunctively treat certain diseases like metabolic and immune disorders. OBJECTIVE We recently generated a space-flight mutated Lactobacillus plantarum SS18-50 with good in vitro probiotic characteristics. In the current research, we designed two in vivo experiments to evaluate whether L. plantarum SS18-50 had the ability to increase beneficial gut bacteria, regulate oxidative status and ameliorate inflammation in mice. METHODS Experiments I: the ICR mice were gavaged with L. plantarum SS18-50 or its wild type L. plantarum GS18 at 107 or 109 CFU/kg BW daily for one month, during which the body weight was recorded weekly. The feces were collected to determine the abundance of two main beneficial bacterial groups including Lactobacillus and Bifidobacterium by selective culturing, while the total triglycerides and cholesterols in sera were determined using commercial kits. Experiment II: the mice were gavaged with loperamide hydrochloride to develop oxidative stress and inflammation phenotypes. At the same time, the experimental mice were gavaged with L. plantarum SS18-50 or wild type L. plantarum GS18 at 107 or 109 CFU/kg BW daily for one month. At the end of experiment, oxidative indicators (SOD and MDA) and inflammatory cytokines (IL-17A and IL-10) were measured by commercial kits. RESULTS Results showed that L. plantarum SS18-50 increased the abundance of Lactobacillus and Bifidobacterium in mice after one month's administration. L. plantarum SS18-50 also showed the anti-oxidant activity by increasing SOD and decreasing MDA, and exerted the anti-inflammatory effect by increasing IL-10 and decreasing IL-17A in Lop treated mice. Both the wild type stain and the space mutant had such biomedical effects, but L. plantarum SS18-50 was better in increasing gut beneficial bacteria and oxidative regulation than the wild type (P<0.05). CONCLUSION we conclude that L. plantarum SS18-50 has a great potential to serve as a dietary functional probiotic supplement and/or adjunctive treatment strategy.
Collapse
Affiliation(s)
- Dan Wang
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Rd., Changchun, China
| | - Tiehua Zhang
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Rd., Changchun, China
| | - Hongwei Hao
- Fullarton Bioengineering Technology Co., Ltd, Beijing, China
| | - Hongxing Zhang
- College of Food Science and Engineering, Beijing University of Agriculture, Beijing, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Rd., Changchun, China
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, 5333 Xi'an Rd., Changchun, China
| |
Collapse
|
6
|
Hajare ST. Effects of potential probiotic strains LBKV-3 on Immune Cells responses in Malnutrite children: a double-blind, randomized, Controlled trial. J Immunoassay Immunochem 2021; 42:453-466. [PMID: 33750266 DOI: 10.1080/15321819.2021.1895217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This study investigated the phagocytic cell response in malnourished children after oral feeding of a fermented product containing clinically proven probiotic strains of Lactobacillus acidophilus, LBKV-3. The bacterial strain is used as a probiotic for humans to test its effect on immune cell activity in undernourished children below 8 years of age. To study the immune cell activity, implantation abilities of the culture in the GI tract of malnourished children, forty-five children of 6-7 and 7-8 years were randomly selected and distributed in three groups, each comprising 15 children in each of the age group. The test group of the children was receiving 100 g product volunteers/day of freshly prepared probiotic acidophilus milk containing 107 cfu/g of culture. The control group was receiving 100 g freshly prepared "dahi" containing 107 cfu/g of the LAB while the blank group of the volunteers was receiving thermal processed (85° C/30 min) buffalo milk containing 5% fat and 10% SNF at the rate of 100 ml/day/volunteer. Feeding trial was continued for 12 weeks. Blood samples were collected at W2, W4, W8, and W12. The blood serum samples were analyzed for monocytes, neutrophils, basophils, and lymphocytes by BC-3000 + Auto Hematology Analyzer. In conclusion, consumption of PAM increased the proportion of immune cells, including monocytes, neutrophils, basophils, and lymphocytes, as well as their phagocytic activity in all age groups but proportion was significant in the test group of 7-8 years. The effects were higher during W12 compared to W2, W4, and W8, which suggest regulation of the immune system.
Collapse
Affiliation(s)
- Sunil T Hajare
- College of Natural and Computational Sciences, Dilla University, Ethiopia
| |
Collapse
|
7
|
Zhang Y, Wu T, Chen Z, Meng Y, Zhu Z, Wang Q, Tian J, Yi D, Wang L, Zhao D, Hou Y. Dietary Supplementation with Enterococcus faecium R1 Attenuates Intestinal and Liver Injury in Piglets Challenged by Lipopolysaccharide. ANIMALS : AN OPEN ACCESS JOURNAL FROM MDPI 2021; 11:ani11051424. [PMID: 34065711 PMCID: PMC8156331 DOI: 10.3390/ani11051424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
Simple Summary The biological characteristics of E. faecium R1 and the effect of dietary supplementation with E. faecium R1 on the growth performance of weaned piglets were studied. The results showed that E. faecium R1 had the characteristics of effective bacteriostatic activity, acid resistance, bile salt resistance, and high-temperature resistance. Dietary supplementation with E. faecium R1 (6.5 × 106 CFU/g) improved intestinal function of weaning piglets by decreasing diarrhea incidence. Further research found that dietary supplementation with E. faecium R1 (6.5 × 106 CFU/g) attenuated intestinal and liver injury in piglets challenged by lipopolysaccharide. Abstract In this study, a strain of E. faecium R1 with effective bacteriostatic activity, acid resistance, bile salt resistance, high-temperature resistance was screened. To study the effect of E. faecium R1 on lipopolysaccharide (LPS)-induced intestinal and liver injury in piglets, twenty-four weaned female piglets were randomly assigned into one of three groups (8 piglets per group). Piglets in the control group and LPS group were fed a basal diet, piglets in the E. faecium group were fed the basal diet supplemented with E. faecium R1 (6.5 × 106 CFU/g). On day 21 of the trial, piglets in the LPS group and E. faecium group were intraperitoneally administered LPS (100 μg/kg), piglets in the control group were administered the same volume of saline. Subsequently, blood samples were collected at 3 h, and intestinal, liver, and pancreas samples were collected at 6 h. Results showed that E. faecium R1 supplementation significantly decreased the diarrhea rate and feed to gain ratio, and dramatically reduced LPS-induced intestinal and liver injury in piglets. Compared with the LPS group, E. faecium R1 supplementation significantly increased the content of glucagon in plasma and IL-1β in the liver, and the mRNA levels of villin in jejunum and ileum and Bcl-xL and pBD-L in the ileum, and significantly decreased the contents of prostaglandin 2 and malondialdehyde in the liver and the activities of myeloperoxidase and aspartate aminotransferase in plasma in piglets. Moreover, E. faecium R1 improved the pancreatic antioxidant capacity in piglets, which was indicated by a significant increase in catalase activity and a decrease in total nitric oxide synthase activity. In summary, dietary supplementation with E. faecium R1 alleviates intestinal and liver injury in LPS-challenged piglets.
Collapse
|
8
|
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
|
9
|
Wang BX, Wu CM, Ribbeck K. Home, sweet home: how mucus accommodates our microbiota. FEBS J 2021; 288:1789-1799. [PMID: 32755014 PMCID: PMC8739745 DOI: 10.1111/febs.15504] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/17/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022]
Abstract
As a natural environment for human-microbiota interactions, healthy mucus houses a remarkably stable and diverse microbial community. Maintaining this microbiota is essential to human health, both to support the commensal bacteria that perform a wide array of beneficial functions and to prevent the outgrowth of pathogens. However, how the host selects and maintains a specialized microbiota remains largely unknown. In this viewpoint, we propose several strategies by which mucus may regulate the composition and function of the human microbiota and discuss how compromised mucus barriers in disease can give rise to microbial dysbiosis.
Collapse
Affiliation(s)
- Benjamin X Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Chloe M Wu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
10
|
Zhang N, Li C, Niu Z, Kang H, Wang M, Zhang B, Tian H. Colonization and immunoregulation of Lactobacillus plantarum BF_15, a novel probiotic strain from the feces of breast-fed infants. Food Funct 2021; 11:3156-3166. [PMID: 32207765 DOI: 10.1039/c9fo02745a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immunosuppression is a manifestation imbalance in the immune system, often during unhealthy states. In recent years, lactic acid bacteria (LAB) have been found to be important components of the body's innate immune system, and indispensable to maintaining normal immune function. Lactobacillus plantarum BF_15, a novel strain isolated from the feces of breast-fed infants, which has shown potential as an immunomodulator in vitro. In the present study, with the Polymerase Chain Reaction-Denaturing Gradient Gel Electrophoresis (PCR-DGGE) based on RNA-polymerase beta subunit encoding gene (rpoB) to analyze the colonization of L. plantarum BF_15 in the intestine of mice. In addition, Lactobacillus rhamnosus GG (LGG) as a positive control strain, by measuring immune-related indexes and the diversity of intestinal microbiota, the effects of BF_15 on immunoregulation and intestinal microbiota dysbiosis were investigated to elucidate whether the attenuation of immunosuppression is related to the modulation of intestinal microbiota. Results did indeed support this notion that BF_15 did colonize murine intestines well, in which it could still be detected in mice feces 14 days after stopping the probiotic administration. Moreover, BF_15 found to protect mice against reduction in the levels of several immune-related indicators, including the thymus and spleen indexes, splenic lymphocyte proliferation, toe swelling degree, serum hemolysin-antibody level, and macrophage phagocytosis index, triggered by high-dose (200 mg kg-1) intraperitoneal administration of cyclophosphamide (CTX). In addition, the strain was also found to effectively balance intestinal microbiota dysbiosis in the mice. Collectively, these results indicated that L. plantarum BF_15 can not only successfully colonize murine intestines, but also can effectively alleviate CTX-induced immunosuppression, once established, by rebalancing the intestinal microbiota. This, therefore, provides strong evidence for the view that BF_15 has the potential to become a highly effective immunomodulating probiotic in human microbiota as well.
Collapse
Affiliation(s)
- Na Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and College of Biochemistry and Environmental Engineering, Baoding University, Baoding, Hebei 071000, China
| | - Chen Li
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Zhihua Niu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongyan Kang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Bo Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongtao Tian
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and National Engineering Research Center for Agriculture in Northern Mountainous Areas, Baoding, Hebei 071000, China
| |
Collapse
|
11
|
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
|
12
|
Tong J, Satyanarayanan SK, Su H. Nutraceuticals and probiotics in the management of psychiatric and neurological disorders: A focus on microbiota-gut-brain-immune axis. Brain Behav Immun 2020; 90:403-419. [PMID: 32889082 DOI: 10.1016/j.bbi.2020.08.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
- Jiaqi Tong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao.
| |
Collapse
|
13
|
Potential Cultivation of Lactobacillus pentosus from Human Breastmilk with Rapid Monitoring through the Spectrophotometer Method. Processes (Basel) 2020. [DOI: 10.3390/pr8080902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The present study focused on the development of a new method to determine the lag phase of Lactobacillus in breast milk which was attained during the 1st, 3rd, and 6th month (M1, M3, and M6). The colonies’ phylogenetic analysis, derived from the 16S rRNA gene sequences, was evaluated with genus Lactobacillus pentosus and achieved a similarity value of 99%. Raman spectroscopy in optical densities of 600 nm (OD600) were used for six consecutive days to observe the changes of the cell growth rate. The values of OD600 were well fitted with the regression model. From this work, M1 was found to be the longest lag phase in 18 h, and it was 17% to 27% longer compared to M3 and M6, respectively. However, the samples of M3 and M6 showed the shortest duration in reaching 0.5 of OD600 nm (16 h) which was enhanced by 80% and 96% compared to M1, respectively. These studies will be of significance when applied in determining the bacteria growth curve and in assessing the growth behavior for the strain in human breast milk.
Collapse
|
14
|
Wu AJ, Lee DJ, Li F, Tobin NH, Aldrovandi GM, Shew SB, Calkins KL. Impact of Clinical Factors on the Intestinal Microbiome in Infants With Gastroschisis. JPEN J Parenter Enteral Nutr 2020; 45:818-825. [PMID: 32441784 DOI: 10.1002/jpen.1926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Infants with gastroschisis require operations and lengthy hospitalizations due to intestinal dysmotility. Dysbiosis may contribute to these problems. Little is known on the microbiome of gastroschisis infants. METHODS The purpose of this study was to investigate the fecal microbiome in gastroschisis infants. Microbiome profiling was performed by sequencing the V4 region of the 16S rRNA gene. The microbiome of gastroschisis infants was compared with the microbiome of healthy controls, and the effects of mode of birth delivery, gestational age, antibiotic duration, and nutrition type on microbial composition and diversity were investigated. RESULTS The microbiome of gastroschisis infants (n = 13) was less diverse (Chao1, P < .001), lacked Bifidobacterium (P = .001), and had increased Staphylococcus (P = .007) compared with controls (n = 83). Mode of delivery (R2 = 0.04, P = .001), antibiotics duration ≥7 days (R2 = 0.03, P = .003), age at sample collection (R2 = 0.03, P = .009), and gestational age (R2 = 0.02, P = .035) explained a small portion of microbiome variation. In gastroschisis infants, Escherichia-Shigella was the predominate genus, and those delivered via cesarean section had different microbial communities, predominantly Staphylococcus and Streptococcus, from those delivered vaginally. Although antibiotic duration contributed to the variation in microbiome composition, there were no significant differences in taxa distribution or α diversity by antibiotic duration or nutrition type. CONCLUSION The microbiome of gastroschisis infants is dysbiotic, and mode of birth delivery, antibiotic duration, and gestational age appear to contribute to microbial variation.
Collapse
Affiliation(s)
- Allison J Wu
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.,Department of Pediatrics, Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David J Lee
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Fan Li
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Nicole H Tobin
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Grace M Aldrovandi
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Stephen B Shew
- Department of Surgery, Division of Pediatric Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kara L Calkins
- Department of Pediatrics, Neonatal Research Center of the UCLA of Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA and UCLA Mattel Children's Hospital, Los Angeles, California, USA
| |
Collapse
|
15
|
Microbiome Composition in Pediatric Populations from Birth to Adolescence: Impact of Diet and Prebiotic and Probiotic Interventions. Dig Dis Sci 2020; 65:706-722. [PMID: 32002758 PMCID: PMC7046124 DOI: 10.1007/s10620-020-06092-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diet is a key regulator of microbiome structure and function across the lifespan. Microbial colonization in the first year of life has been actively researched; however, studies during childhood are sparse. Herein, the impact of dietary intake and pre- and probiotic interventions on microbiome composition of healthy infants and children from birth to adolescence is discussed. The microbiome of breastfed infants has lower microbial diversity and richness, higher Proteobacteria, and lower Bacteroidetes and Firmicutes than those formula-fed. As children consume more complex diets, associations between dietary patterns and the microbiota emerge. Like adults, the microbiota of children consuming a Western-style diet is associated with greater Bacteroidaceae and Ruminococcaceae and lower Prevotellaceae. Dietary fibers and pre- or/and probiotics have been tested to modulate the gut microbiota in early life. Human milk oligosaccharides and prebiotics added to infant formula are bifidogenic and decrease pathogens. In children, prebiotics, such as inulin, increase Bifidobacterium abundance and dietary fibers reduce fecal pH and increase alpha diversity and calcium absorption. Probiotics have been administered to the mother during pregnancy and breastfeeding or directly to the infant/child. Findings on maternal probiotic administration on bacterial taxa are inconsistent. When given directly to the infant/child, some changes in individual taxa are observed, but rarely is overall alpha or beta diversity affected. Cesarean-delivered infants appear to benefit to a greater degree than those born vaginally. Infancy and childhood represent an opportunity to beneficially manipulate the microbiome through dietary or prebiotic interventions, which has the potential to affect both short- and long-term health outcomes.
Collapse
|
16
|
Kumar M, Singh P, Murugesan S, Vetizou M, McCulloch J, Badger JH, Trinchieri G, Al Khodor S. Microbiome as an Immunological Modifier. Methods Mol Biol 2020; 2055:595-638. [PMID: 31502171 PMCID: PMC8276114 DOI: 10.1007/978-1-4939-9773-2_27] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Humans are living ecosystems composed of human cells and microbes. The microbiome is the collection of microbes (microbiota) and their genes. Recent breakthroughs in the high-throughput sequencing technologies have made it possible for us to understand the composition of the human microbiome. Launched by the National Institutes of Health in USA, the human microbiome project indicated that our bodies harbor a wide array of microbes, specific to each body site with interpersonal and intrapersonal variabilities. Numerous studies have indicated that several factors influence the development of the microbiome including genetics, diet, use of antibiotics, and lifestyle, among others. The microbiome and its mediators are in a continuous cross talk with the host immune system; hence, any imbalance on one side is reflected on the other. Dysbiosis (microbiota imbalance) was shown in many diseases and pathological conditions such as inflammatory bowel disease, celiac disease, multiple sclerosis, rheumatoid arthritis, asthma, diabetes, and cancer. The microbial composition mirrors inflammation variations in certain disease conditions, within various stages of the same disease; hence, it has the potential to be used as a biomarker.
Collapse
Affiliation(s)
- Manoj Kumar
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Parul Singh
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Selvasankar Murugesan
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Marie Vetizou
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John McCulloch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Souhaila Al Khodor
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
17
|
Van Daele E, Knol J, Belzer C. Microbial transmission from mother to child: improving infant intestinal microbiota development by identifying the obstacles. Crit Rev Microbiol 2019; 45:613-648. [DOI: 10.1080/1040841x.2019.1680601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Emmy Van Daele
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Gut Biology and Microbiology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
18
|
Nie P, Li Z, Wang Y, Zhang Y, Zhao M, Luo J, Du S, Deng Z, Chen J, Wang Y, Chen S, Wang L. Gut microbiome interventions in human health and diseases. Med Res Rev 2019; 39:2286-2313. [PMID: 30994937 DOI: 10.1002/med.21584] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/27/2019] [Accepted: 03/24/2019] [Indexed: 02/06/2023]
Abstract
Ongoing studies have determined that the gut microbiota is a major factor influencing both health and disease. Host genetic factors and environmental factors contribute to differences in gut microbiota composition and function. Intestinal dysbiosis is a cause or a contributory cause for diseases in multiple body systems, ranging from the digestive system to the immune, cardiovascular, respiratory, and even nervous system. Investigation of pathogenesis has identified specific species or strains, bacterial genes, and metabolites that play roles in certain diseases and represent potential drug targets. As research progresses, gut microbiome-based diagnosis and therapy are proposed and applied, which might lead to considerable progress in precision medicine. We further discuss the limitations of current studies and potential solutions.
Collapse
Affiliation(s)
- Pengqing Nie
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yimeng Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yubing Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Mengna Zhao
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shiming Du
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zixin Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Jincao Chen
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yunfu Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shi Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Lianrong Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
19
|
McKeen S, Young W, Mullaney J, Fraser K, McNabb WC, Roy NC. Infant Complementary Feeding of Prebiotics for theMicrobiome and Immunity. Nutrients 2019; 11:nu11020364. [PMID: 30744134 PMCID: PMC6412789 DOI: 10.3390/nu11020364] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Complementary feeding transitions infants from a milk-based diet to solid foods, providing essential nutrients to the infant and the developing gut microbiome while influencing immune development. Some of the earliest microbial colonisers readily ferment select oligosaccharides, influencing the ongoing establishment of the microbiome. Non-digestible oligosaccharides in prebiotic-supplemented formula and human milk oligosaccharides promote commensal immune-modulating bacteria such as Bifidobacterium, which decrease in abundance during weaning. Incorporating complex, bifidogenic, non-digestible carbohydrates during the transition to solid foods may present an opportunity to feed commensal bacteria and promote balanced concentrations of beneficial short chain fatty acid concentrations and vitamins that support gut barrier maturation and immunity throughout the complementary feeding window.
Collapse
Affiliation(s)
- Starin McKeen
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Wayne Young
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Jane Mullaney
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Karl Fraser
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Nicole C Roy
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
20
|
Tsunoda Y, Asahara T, Nomoto K, Yoshioka Y, Fukuma E. Bacterial profile of infant feces associated with lactation infectious breasts. PEDIATRIC HEALTH MEDICINE AND THERAPEUTICS 2018; 9:173-180. [PMID: 30584388 PMCID: PMC6287549 DOI: 10.2147/phmt.s184637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background Mastitis is a common complication in lactating women. However, the diversity of intestinal bacteria in infant exclusively fed infectious milk remains uncharacterized. Our colleagues recently established a method based on 16S and 23S rRNA-targeted reverse transcription–quantitative PCR (RT-qPCR) for detecting bacteria. Materials and methods In the present study, the bacteria present in 14 samples of milk and infant feces were characterized using the RT-qPCR method, and concentrations of fecal organic acids were measured during the period of breast massage using HPLC. Results Streptococcus agalactiae and Str. parasanguinis were detected in milk from mastitis patients, whereas Str. salivarius and Str. thermophilus were the predominant bacteria in milk from engorged breasts. In feces of breastfed infants, Str. salivarius, Str. thermophiles, and Str. parasanguinis were isolated. Levels of lactate were high in fecal samples, whereas the pH of infant feces stabilized during breast massage. The bacterial diversity of milk from lactation infectious breasts was similar to that in feces of infant fed milk from lactation infectious breasts. Streptococcus species isolated from the feces of breastfed infants are related to oral cavity health. Conclusion These results suggest that Streptococcus species, which are part of the healthy oral microflora, may play an important role in preserving the intestinal bacterial flora in infants fed infectious milk.
Collapse
Affiliation(s)
- Yuko Tsunoda
- Department of Breast Surgery, Breast Center, Kameda Medical Center, Kamogawa, Chiba 296-8602, Japan,
| | - Takashi Asahara
- Department of Microbiological Research, Yakult Central Institute, Kunitachi, Tokyo 186-8650, Japan
| | - Koji Nomoto
- Department of Molecular Microbiology, Tokyo University of Agriculture, Setagaya-ku, Tokyo 156-8502, Japan
| | - Yukiko Yoshioka
- Department of Breast Management Research, Oketani Breast Management Research Institute, Shinjuku-ku, Tokyo 162-0044, Japan
| | - Eisuke Fukuma
- Department of Breast Surgery, Breast Center, Kameda Medical Center, Kamogawa, Chiba 296-8602, Japan,
| |
Collapse
|
21
|
Mohammadkhah AI, Simpson EB, Patterson SG, Ferguson JF. Development of the Gut Microbiome in Children, and Lifetime Implications for Obesity and Cardiometabolic Disease. CHILDREN (BASEL, SWITZERLAND) 2018; 5:E160. [PMID: 30486462 PMCID: PMC6306821 DOI: 10.3390/children5120160] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/19/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023]
Abstract
Emerging evidence suggests that microbiome composition and function is associated with development of obesity and metabolic disease. Microbial colonization expands rapidly following birth, and microbiome composition is particularly variable during infancy. Factors that influence the formation of the gut microbiome during infancy and childhood may have a significant impact on development of obesity and metabolic dysfunction, with life-long consequences. In this review, we examine the determinants of gut microbiome composition during infancy and childhood, and evaluate the potential impact on obesity and cardiometabolic risk.
Collapse
Affiliation(s)
- Anica I Mohammadkhah
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Eoin B Simpson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Stephanie G Patterson
- Division of Critical Care Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
22
|
Iozzo P, Sanguinetti E. Early Dietary Patterns and Microbiota Development: Still a Way to Go from Descriptive Interactions to Health-Relevant Solutions. Front Nutr 2018; 5:5. [PMID: 29457002 PMCID: PMC5801286 DOI: 10.3389/fnut.2018.00005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/16/2018] [Indexed: 11/13/2022] Open
Abstract
Early nutrition and growth in the initial years of life are important determinants of later body weight and metabolic health in humans, and the current epidemic of obesity involving children requires a better understanding of causal and protective mechanisms and components in infant foods. This review focuses on recent evidence implicating feeding modes (e.g., breast milk and formula milk) and dietary transitions toward complementary foods in the progression of microbiota maturation in children. The literature exploring body weight outcomes of microbiota changes induced by diet in early life is limited. Representative studies addressing the use of probiotics in pregnant women and infants are also examined. Methodological and geo-cultural variations make it difficult to avoid (apparently) controversial findings. Most studies indicate differences in the microbiota of formula versus breastfed infants, but some do not. Duration of breastfeeding delays the maturation of the microbiota toward an adult-like profile. However, the effect size of the early feeding pattern on microbial function was found to be very small, and absent after the third year of life. There are several interesting mediators whereby milk composition can affect infants' microbiota and their optimization is a desirable strategy for prevention. But prevention of what? Although there are few correlative evaluations relating microbiota and body weight in early life, studies demonstrating a cause-effect relationship between diet-induced changes in early microbiota development and subsequent metabolic health outcomes in humans are still missing.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Elena Sanguinetti
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| |
Collapse
|
23
|
Turroni F, Milani C, Duranti S, Ferrario C, Lugli GA, Mancabelli L, van Sinderen D, Ventura M. Bifidobacteria and the infant gut: an example of co-evolution and natural selection. Cell Mol Life Sci 2018; 75:103-118. [PMID: 28983638 PMCID: PMC11105234 DOI: 10.1007/s00018-017-2672-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Throughout the human life, the gut microbiota interacts with us in a number of different ways, thereby influencing our health status. The acquisition of such an interactive gut microbiota commences at birth. Medical and environmental factors including diet, antibiotic exposure and mode of delivery are major factors that shape the composition of the microbial communities in the infant gut. Among the most abundant members of the infant microbiota are species belonging to the Bifidobacterium genus, which are believed to confer beneficial effects upon their host. Bifidobacteria may be acquired directly from the mother by vertical transmission and their persistence in the infant gut is associated with their saccharolytic activity toward glycans that are abundant in the infant gut. Here, we discuss the establishment of the infant gut microbiota and the contribution of bifidobacteria to this early life microbial consortium.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Ferrario
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
24
|
Bifidobacterium bifidum OLB6378 Simultaneously Enhances Systemic and Mucosal Humoral Immunity in Low Birth Weight Infants: A Non-Randomized Study. Nutrients 2017; 9:nu9030195. [PMID: 28245626 PMCID: PMC5372858 DOI: 10.3390/nu9030195] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/10/2017] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Probiotic supplementation has been part of the discussion on methods to enhance humoral immunity. Administration of Bifidobacterium bifidum OLB6378 (OLB6378) reduced the incidence of late-onset sepsis in infants. In this non-randomized study, we aimed to determine the effect of administration of live OLB6378 on infants' humoral immunity. Secondly, we tried to elucidate whether similar effects would be observed with administration of non-live OLB6378. Low birth weight (LBW) infants weighing 1500-2500 g were divided into three groups: Group N (no intervention), Group L (administered live OLB6378 concentrate), and Group H (administered non-live OLB6378 concentrate). The interventions were started within 48 h after birth and continued until six months of age. Serum immunoglobulin G (IgG) levels (IgG at one month/IgG at birth) were significantly higher in Group L than in Group N (p < 0.01). Group H exhibited significantly higher serum IgG levels (p < 0.01) at one month of age and significantly higher intestinal secretory immunoglobulin A (SIgA) levels (p < 0.05) at one and two months of age than Group N. No difference was observed in the mortality or morbidity between groups. Thus, OLB6378 administration in LBW infants enhanced humoral immunity, and non-live OLB6378, which is more useful as a food ingredient, showed a more marked effect than the viable bacteria.
Collapse
|
25
|
Davis EC, Wang M, Donovan SM. The role of early life nutrition in the establishment of gastrointestinal microbial composition and function. Gut Microbes 2017; 8:143-171. [PMID: 28068209 PMCID: PMC5390825 DOI: 10.1080/19490976.2016.1278104] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The development of the human infant intestinal microbiota is a sequential process that begins in utero and continues during the first 2 to 3 years of life. Microbial composition and diversity are shaped by host genetics and multiple environmental factors, of which diet is a principal contributor. An understanding of this process is of clinical importance as the microbiota acquired in early life influence gastrointestinal, immune and neural development, and reduced microbial diversity or dysbiosis during infancy is associated with disorders in infancy and later childhood. The goal of this article was to review the published literature that used culture-independent methods to describe the development of the gastrointestinal microbiota in breast- and formula-fed human infants as well as the impact of prebiotic and probiotic addition to infant formula, and the addition of solid foods.
Collapse
Affiliation(s)
- Erin C. Davis
- Division of Nutritional Sciences, Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA
| | - Sharon M. Donovan
- Division of Nutritional Sciences, Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA,Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL USA,CONTACT Sharon M. Donovan University of Illinois, Department of Food Science and Human Nutrition, 339 Bevier Hall, 905 S. Goodwin Avenue, Urbana, IL 61801
| |
Collapse
|
26
|
Homayoni Rad A, Vaghef Mehrabany E, Alipoor B, Vaghef Mehrabany L. The Comparison of Food and Supplement as Probiotic Delivery Vehicles. Crit Rev Food Sci Nutr 2017; 56:896-909. [PMID: 25117939 DOI: 10.1080/10408398.2012.733894] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Probiotics are live bacteria which have frequently been reported to be beneficial in preventing a wide range of diseases as well as playing a major role in treating the existing ailments. Thus far, a variety of probiotic products have been developed which can be categorized into two groups: probiotic foods and supplements. Both foods and supplements have been able to confer the health benefits claimed for them. However, it is not known which one can be clinically more efficient, and to the best of our knowledge, until now no research has been conducted to investigate this issue. The present review aims to discuss this matter, based on the evidence available in the literature. To do so, articles indexed in PubMed and ScienceDirect between 2000 and 2011 were reviewed. The articles included the clinical trials in which either foods or supplements were used to administer the probiotics to either patients suffering from different diseases or healthy subjects. Although both foods and supplements seem to have been efficient carriers for the beneficial bacteria, to generally promote public health in communities, probiotic foods appear to be preferred to probiotic supplements.
Collapse
Affiliation(s)
- Aziz Homayoni Rad
- a Faculty of Nutrition, Department of Food Science and Technology, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Elnaz Vaghef Mehrabany
- b Faculty of Nutrition, Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Beitullah Alipoor
- b Faculty of Nutrition, Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Leila Vaghef Mehrabany
- c School of Nutritional Sciences and Dietetics, Department of Clinical Nutrition, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
27
|
Abstract
Although largely deprived from exogenous stimuli in utero, the mucosal barriers of the neonate after birth are bombarded by environmental, nutritional, and microbial exposures. The microbiome is established concurrently with the developing immune system. The nature and timing of discrete interactions between these two factors underpins the long-term immune characteristics of these organs, and can set an individual on a trajectory towards or away from disease. Microbial exposures in the gastrointestinal and respiratory tracts are some of the key determinants of the overall immune tone at these mucosal barriers and represent a leading target for future intervention strategies. In this review, we discuss immune maturation in the gut and lung and how microbes have a central role in this process.
Collapse
|
28
|
Dissanayake E, Shimojo N. Probiotics and Prebiotics in the Prevention and Treatment of Atopic Dermatitis. PEDIATRIC ALLERGY, IMMUNOLOGY, AND PULMONOLOGY 2016; 29:174-180. [DOI: 10.1089/ped.2016.0708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Eishika Dissanayake
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoki Shimojo
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
29
|
Ai C, Ma N, Zhang Q, Wang G, Liu X, Tian F, Chen P, Chen W. Immunomodulatory Effects of Different Lactic Acid Bacteria on Allergic Response and Its Relationship with In Vitro Properties. PLoS One 2016; 11:e0164697. [PMID: 27764153 PMCID: PMC5072832 DOI: 10.1371/journal.pone.0164697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023] Open
Abstract
Some studies reported that probiotic could relieve allergy-induced damage to the host, but how to get a useful probiotic is still a challenge. In this study, the protective effects of three lactic acid bacteria (La, Lp and Lc) were evaluated in a mouse model, and its relationship with the in vitro properties was analyzed. The in vitro results indicated that La with the capacity to inhibit IL-4 production could have a better anti-allergy effect in vivo than two others. However, the animal trials showed that all LAB strains could alleviate allergen-induced airway inflammation. Among them, LAB strain Lp had a better effect in inhibiting allergic response through a modulation of Th1/Th2 balance and an increase of regulatory T cells. This difference could be explained by that different LAB strains have a strain-specific effect on gut microbiota closely associated with host immune responses. Finally, this study did not only obtain an effective anti-allergy probiotic strain via animal study, but also indicate that probiotic-induced effect on intestinal microbiota should be considered as an important screening index, apart from its inherent characteristics.
Collapse
Affiliation(s)
- Chunqing Ai
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Na Ma
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- * E-mail: (QXZ); (WC)
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
| | - Pei Chen
- Shaanxi University of Technology, School of Biological Science and Engineering, Hanzhong 723001, P. R. China
- Shanxi Radio & TV University, Xi'an 710119, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, P. R. China
- * E-mail: (QXZ); (WC)
| |
Collapse
|
30
|
Bian G, Ma S, Zhu Z, Su Y, Zoetendal EG, Mackie R, Liu J, Mu C, Huang R, Smidt H, Zhu W. Age, introduction of solid feed and weaning are more important determinants of gut bacterial succession in piglets than breed and nursing mother as revealed by a reciprocal cross-fostering model. Environ Microbiol 2016; 18:1566-77. [DOI: 10.1111/1462-2920.13272] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/17/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Gaorui Bian
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Shouqing Ma
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Zhigang Zhu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Erwin G. Zoetendal
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Roderick Mackie
- Department of Animal Sciences; University of Illinois at Urbana-Champaign; IL USA
| | - Junhua Liu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Chunlong Mu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Ruihua Huang
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Hauke Smidt
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| |
Collapse
|
31
|
Abstract
Toll-like receptor (TLR) signaling is central to innate immunity. Aberrant expression of TLRs is found in neonatal inflammatory diseases. Several bioactive components of human milk modulate TLR expression and signaling pathways, including soluble toll-like receptors (sTLRs), soluble cluster of differentiation (sCD) 14, glycoproteins, small peptides, and oligosaccharides. Some milk components, such as sialyl (α2,3) lactose and lacto-N-fucopentaose III, are reported to increase TLR signaling; under some circumstances this might contribute toward immunologic balance. Human milk on the whole is strongly anti-inflammatory, and contains abundant components that depress TLR signaling pathways: sTLR2 and sCD14 inhibit TLR2 signaling; sCD14, lactadherin, lactoferrin, and 2'-fucosyllactose attenuate TLR4 signaling; 3'-galactosyllactose inhibits TLR3 signaling, and β-defensin 2 inhibits TLR7 signaling. Feeding human milk to neonates decreases their risk of sepsis and necrotizing enterocolitis. Thus, the TLR regulatory components found in human milk hold promise as benign oral prophylactic and therapeutic treatments for the many gastrointestinal inflammatory disorders mediated by abnormal TLR signaling.
Collapse
Affiliation(s)
- YingYing He
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| | - Nathan T Lawlor
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| | - David S Newburg
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA
| |
Collapse
|
32
|
Gollwitzer ES, Marsland BJ. Impact of Early-Life Exposures on Immune Maturation and Susceptibility to Disease. Trends Immunol 2015; 36:684-696. [DOI: 10.1016/j.it.2015.09.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 12/16/2022]
|
33
|
Tewari VV, Dubey SK, Gupta G. Bacillus clausii for Prevention of Late-onset Sepsis in Preterm Infants: A Randomized Controlled Trial. J Trop Pediatr 2015; 61:377-85. [PMID: 26246087 DOI: 10.1093/tropej/fmv050] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND OF THE STUDY Preterm infants are managed with antibiotics for sepsis, including suspected or probable sepsis. This leads to a delayed and abnormal colonization of the gut with potentially pathogenic organisms and a microbiome, which lacks biodiversity and increases the risk for late-onset sepsis (LOS). Probiotics have been proven to reduce the risk for necrotizing enterocolitis, but evidence for prevention of LOS is inconclusive. Probiotic effect depends also on the strain used, dose and indication for use. This study evaluated Bacillus clausii probiotic administered prophylactically to preterm neonates for prevention of LOS. OBJECTIVES To study B.clausii given prophylactically to preterm neonates for prevention of LOS. DESIGN Double-blinded, placebo-controlled, randomized trial. SETTINGS Tertiary care neonatal unit in India. PARTICIPANTS Consecutive preterm neonates <34 weeks, admitted from 1 March 2012 to 28 February 2014 were stratified as extreme preterm and very preterm. INTERVENTION Randomized to receive either probiotic or placebo for 6 weeks, discharge from hospital, death or occurrence of sepsis, whichever was earlier. PRIMARY OUTCOME Incidence of definite and probable LOS in probiotic group compared with placebo. RESULTS Of 326 eligible preterm infants, 244 were enrolled and 82 were excluded. Of these, 120 were stratified as extreme preterm and randomized to receive placebo (n = 59) and probiotic (n = 61). Of 124 babies stratified as very preterm, an equal number was randomized to receive placebo (n = 62) and probiotic (n = 62). There was no significant difference in the incidence of LOS between the two arms in the extreme preterm group [29% vs. 23%; relative risk (RR) 1.27; 95% confidence interval (CI) 0.88-1.66; p = 0.36] and the very preterm group (13% vs. 10%; RR 1.33; 95% CI 0.96-1.70; p = 0.32). Full feeds were achieved significantly faster in the probiotic group in both the extreme preterm (RR 0.82; 95% CI 0.74-0.88) and the very preterm (RR 0.67; 95% 0.32-0.77). CONCLUSIONS Prophylactic administration of B.clausii to preterm neonates did not result in a significant difference in the incidence of LOS as compared with placebo.
Collapse
Affiliation(s)
| | - Sachin Kumar Dubey
- Department of Pediatrics, Army Hospital (Referral & Research) New Delhi, India
| | | |
Collapse
|
34
|
Taghizadeh M, Mirlohi M, Poursina F, Madani G, Khoshhali M, Bahreini N, Safaei HG. The influence of impact delivery mode, lactation time, infant gender, maternal age and rural or urban life on total number of Lactobacillus in breast milk Isfahan - Iran. Adv Biomed Res 2015; 4:141. [PMID: 26322289 PMCID: PMC4549918 DOI: 10.4103/2277-9175.161546] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/06/2015] [Indexed: 12/13/2022] Open
Abstract
Background: Breast milk is known as the most crucial postpartum issue in metabolic and immunologic programming of neonatal health. Human milk microbial changes over Lactation. The factors influencing the milk microbiome as well as potential impact of microbes on infant health have not yet been discovered. The objective was to identify pre- and post-natal factors that can potentially influence the bacterial communities inhabiting human milk. Materials and Methods: Breast milk samples (n = 40) with all full-term breastfed infants were collected from lactating randomized. Information on personal characteristics, dietary habits, information about infants were collected after birth. The samples were plated with serial dilutions on three selective culture media man rogosa sharp and then colonies were counted. Colonies tested for catalase reaction, Gram-staining and microscopic examination. Results: The result of this study showed that the overall incidence of positive Lactobacillus in mother's milk was 87.5%. The results based on (infant gender, mode of delivery, rural or urban and lactation time) rural or urban and lactation time were significant (P < 0.05). The results showed that all of the variables were significant in this regression model (P < 0.001). The median of log10Lactobacillus counts in rural mothers, vaginal delivery, infant male gender and Lactation time for first 3-month were meaningfully high. Conclusions: The findings of this study about the breast milk Lactobacillus potential probiotic bacteria of healthy Iranian mothers, suggested that the breast milk microbiome is significantly influenced by several factors, mode of delivery, rural or urban and lactation time.
Collapse
Affiliation(s)
- Mansoureh Taghizadeh
- Department of Food Science and Technology, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Mirlohi
- Department of Food Science and Technology, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farkhondeh Poursina
- Department of Microbiology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnoush Madani
- Department of Food Science and Technology, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehri Khoshhali
- Department of Biostatistics and Epidemiology, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nimah Bahreini
- Department of Food Science and Technology, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hajieh Ghasemian Safaei
- Food Security Research Center, Department of Microbiology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Scharek-Tedin L, Kreuzer-Redmer S, Twardziok SO, Siepert B, Klopfleisch R, Tedin K, Zentek J, Pieper R. Probiotic Treatment Decreases the Number of CD14-Expressing Cells in Porcine Milk Which Correlates with Several Intestinal Immune Parameters in the Piglets. Front Immunol 2015; 6:108. [PMID: 25806034 PMCID: PMC4354412 DOI: 10.3389/fimmu.2015.00108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/25/2015] [Indexed: 11/13/2022] Open
Abstract
Modulating the mucosal immune system of neonates by probiotic treatment of their mothers is a promising approach which can only be investigated through the use of animal models. Here, we used sows and their piglets to investigate the impact of a bacterial treatment on the sow's milk and on the neonate piglet intestinal immune system. In previous experiments, feed supplementation of sows with the probiotic Enterococcus faecium NCIMB 10415 during pregnancy and lactation had been shown to affect intestinal microbiota and cytokine expression of the offspring during the suckling and weaning periods. We therefore investigated the composition of the milk from treated sows in comparison to samples from a control group. In treated sows, the amount of lactose increased, and the somatic cell numbers were reduced. In all milk samples, the percentage of cells expressing membranous CD14 (mCD14) was greater than the fractions of immune cells, indicating expression of mCD14 on mammary epithelial cells. However, in the milk of E. faecium-treated sows, mCD14(+) cells were reduced. Furthermore, the number of CD14(+) milk cells was positively correlated with the percentages of B cells and activated T cells in the ileal MLN of the piglets. This study provides evidence for the expression of mCD14 by the porcine mammary epithelium, and suggests an immunological effect of mCD14(+) milk cells on the piglets' intestinal immune system. Our study further suggests that mCD14(+) mammary epithelial cell populations can be modulated by probiotic feed supplementation of the sow.
Collapse
Affiliation(s)
| | - Susanne Kreuzer-Redmer
- Breeding Biology and Molecular Genetics, Humboldt-Universität zu Berlin , Berlin , Germany
| | - Sven Olaf Twardziok
- Institute of Molecular Biology and Bioinformatics, Charité-Universitätsmedizin Berlin , Berlin , Germany
| | - Bianca Siepert
- Institute of Microbiology and Epizootics, Freie Universität Berlin , Berlin , Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin , Berlin , Germany
| | - Karsten Tedin
- Institute of Microbiology and Epizootics, Freie Universität Berlin , Berlin , Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin , Berlin , Germany
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin , Berlin , Germany
| |
Collapse
|
36
|
Human milk and infant intestinal mucosal glycans guide succession of the neonatal intestinal microbiota. Pediatr Res 2015; 77:115-20. [PMID: 25356747 DOI: 10.1038/pr.2014.178] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Infants begin acquiring intestinal microbiota at parturition. Initial colonization by pioneer bacteria is followed by active succession toward a dynamic ecosystem. Keystone microbes engage in reciprocal transkingdom communication with the host, which is essential for human homeostasis and health; therefore, these bacteria should be considered mutualists rather than commensals. This review discusses the maternal role in providing infants with functional and stable microbiota. The initial fecal inoculum of microbiota results from the proximity of the birth canal and anus; the biological significance of this anatomic proximity could underlie observed differences in microbiota between vaginal and cesarean birth. Secondary sources of inocula include mouths and skin of kin, animals and objects, and the human milk microbiome, but guiding microbial succession may be a primary role of human milk. The unique glycans of human milk cannot be digested by the infant, but are utilized by mutualist bacteria. These prebiotic glycans support expansion of mutualist microbiota, which manifests as differences in microbiota among breastfed and artificially fed infants. Human milk glycans vary by maternal genotype. Milks of genetically distinct mothers and variations in infant mucosal glycan expression support discrete microbiota. Early colonization may permanently influence microbiota composition and function, with ramifications for health.
Collapse
|
37
|
Ahn J. Physicochemical, Microbial, and Sensory Properties of Yogurt with Ulmus davidiana var. japonica During Storage. ACTA ACUST UNITED AC 2014. [DOI: 10.7856/kjcls.2014.25.4.601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Abstract
Enteric viral infections in domestic animals cause significant economic losses. The recent emergence of virulent enteric coronaviruses [porcine epidemic diarrhea virus (PEDV)] in North America and Asia, for which no vaccines are available, remains a challenge for the global swine industry. Vaccination strategies against rotavirus and coronavirus (transmissible gastroenteritis virus) infections are reviewed. These vaccination principles are applicable against emerging enteric infections such as PEDV. Maternal vaccines to induce lactogenic immunity, and their transmission to suckling neonates via colostrum and milk, are critical for early passive protection. Subsequently, in weaned animals, oral vaccines incorporating novel mucosal adjuvants (e.g., vitamin A, probiotics) may provide active protection when maternal immunity wanes. Understanding intestinal and systemic immune responses to experimental rotavirus and transmissible gastroenteritis virus vaccines and infection in pigs provides a basis and model for the development of safe and effective vaccines for young animals and children against established and emerging enteric infections.
Collapse
Affiliation(s)
- Kuldeep S Chattha
- Canadian Food Inspection Agency, Lethbridge, Alberta T1H 6P7, Canada;
| | | | | |
Collapse
|
39
|
Prevention and Control of Diseases by Use of Pro- and Prebiotics (Synbiotics). FOOD REVIEWS INTERNATIONAL 2014. [DOI: 10.1080/87559129.2014.929142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Human milk composition differs in healthy mothers and mothers with celiac disease. Eur J Nutr 2014; 54:119-28. [DOI: 10.1007/s00394-014-0692-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/20/2014] [Indexed: 12/12/2022]
|
41
|
Impact of orally administered lozenges with Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 on the number of salivary mutans streptococci, amount of plaque, gingival inflammation and the oral microbiome in healthy adults. Clin Oral Investig 2014; 19:77-83. [PMID: 24638207 PMCID: PMC4286650 DOI: 10.1007/s00784-014-1221-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/26/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The aim was to evaluate the effects of orally administered Lactobacillus rhamnosus GG (LGG) and Bifidobacterium animalis subsp. lactis BB-12 (BB-12) on the number of salivary mutans streptococci (MS), amount of plaque, gingival inflammation and the oral microbiota in healthy young adults. MATERIALS AND METHODS The study was a randomised, controlled, double-blind trial. Healthy volunteers used lozenges containing a combination of LGG and BB-12 (test group, n = 29) or lozenges without added probiotics (control group, n = 31) for 4 weeks. At baseline and at the end of the test period, the plaque index (PI) and gingival index (GI) were determined, and stimulated saliva was collected. The microbial composition of saliva was assessed using human oral microbe identification microarray (n = 30). MS and lactobacilli (LB) were plate cultured. RESULTS The probiotic lozenge decreased both PI and GI (p < 0.05) while no changes were observed in the control group. However, no probiotic-induced changes were found in the microbial compositions of saliva in either group. CONCLUSIONS The probiotic lozenge improved the periodontal status without affecting the oral microbiota. CLINICAL RELEVANCE Short-term consumption of LGG and BB-12 decreased the amount of plaque which was associated with a clinical impact: a decrease in gingival inflammation.
Collapse
|
42
|
Shafi A, Farooq U, Akram K, Jaskani M, Siddique F, Tanveer A. Antidiarrheal Effect of Food Fermented by Various Strains ofLactobacillus. Compr Rev Food Sci Food Saf 2014; 13:229-239. [DOI: 10.1111/1541-4337.12056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/23/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Afshan Shafi
- Inst. of Food Science and Nutrition; Univ. of Sargodha; Sargodha Pakistan
| | - Umar Farooq
- Inst. of Food Science and Nutrition; Univ. of Sargodha; Sargodha Pakistan
| | - Kashif Akram
- Inst. of Food Science and Nutrition; Univ. of Sargodha; Sargodha Pakistan
| | - Mahgul Jaskani
- Inst. of Food Science and Nutrition; Univ. of Sargodha; Sargodha Pakistan
| | - Farzana Siddique
- Dept. of Food Technology; Pir Mehr Ali Shah Arid Agriculture Univ.; Rawalpindi Pakistan
| | - Amna Tanveer
- Inst. of Food Science and Nutrition; Univ. of Sargodha; Sargodha Pakistan
| |
Collapse
|
43
|
Nikniaz L, Ostadrahimi A, Mahdavi R, Hejazi MA, Salekdeh GH. Effects of synbiotic supplementation on breast milk levels of IgA, TGF-β1, and TGF-β2. J Hum Lact 2013; 29:591-6. [PMID: 23788632 DOI: 10.1177/0890334413490833] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Effects of probiotics on the immunological composition of breast milk have been investigated in a few previous studies. OBJECTIVES The aims of this study were to determine the effects of synbiotic (probiotic plus prebiotic) supplementation on immunoglobulin A (IgA), transforming growth factor β1 (TGF-β1), and transforming growth factor β2 (TGF-β2) levels of breast milk and on diarrhea incidence in infants. METHODS In this randomized, double-blind, and placebo-controlled trial, we recruited 80 lactating mothers who were exclusively breastfeeding their 3-month-old infants. We randomly divided the mothers into 2 groups to receive a daily synbiotic supplement (n = 40) or a placebo (n = 40) for 30 days. Demographic and clinical data (ie, health status) were obtained through an interview. The IgA levels of breast milk were detected by nephelometry, and the levels of TGF-β1 and TGF-β2 were measured using a commercial Platinum ELISA kit. RESULTS The breast milk IgA increased significantly from 0.41 ± 0.09 to 0.48 ± 0.15 g/L in the supplemented group (P = .018), while in the placebo group, no significant changes were observed. Although the breast milk TGF-β1 levels did not change significantly, the TGF-β2 levels of breast milk increased significantly from 270 ± 37.8 to 382 ± 43.7 pg/mL in the supplemented group (P = .043). Also, the incidence of diarrhea in infants decreased significantly in the supplemented group while no significant changes were observed in the placebo group after the experimental period. CONCLUSION Synbiotic supplementation may have positive effects on the immune composition of breast milk and the reduction of diarrhea incidence in infants.
Collapse
Affiliation(s)
- Leila Nikniaz
- 1Student Research Committee, School of Public Health & Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | |
Collapse
|
44
|
Perinatal outcomes of prenatal probiotic and prebiotic administration: an integrative review. J Perinat Neonatal Nurs 2013; 27:288-301; quiz E1-2. [PMID: 24164813 DOI: 10.1097/jpn.0b013e3182a1e15d] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The purpose of this integrative review was to identify, critique, and synthesize the maternal and neonatal evidence on the prenatal use of probiotics and prebiotics to inform perinatal health professionals. A comprehensive literature search resulted in 37 studies of prenatal probiotics and 1 on antepartal prebiotics published from 1990 through 2011 that reported maternal, fetal, and/or neonatal outcomes. The methodologic quality of the studies reviewed was high, although investigators used different probiotic combinations and inconsistently reported perinatal clinical outcomes. The extraction of perinatal outcome variables resulted in identification of 9 maternal and 5 neonatal categories. Prenatal probiotics significantly reduced the incidence of bacterial vaginosis, increased colonization with vaginal Lactobacillus and intestinal Lactobacillus rhamnosus, altered immune markers in serum and breast milk, improved maternal glucose metabolism, and reduced the incidence of gestational diabetes and preeclampsia. Antepartally, probiotics were associated with significantly higher counts of Bifidobacterium and Lactococcus lactis (healthy intestinal flora) in neonatal stool. Prenatal prebiotics significantly increased maternal intestinal Bifidobacterium. No adverse events were reported and there was evidence of safety and tolerance of prenatal probiotics and prebiotics in the scientific investigations reviewed. It is recommended that in future investigations of prenatal probiotics researchers explicitly report maternal and neonatal outcomes.
Collapse
|
45
|
Spencer SJ. Perinatal nutrition programs neuroimmune function long-term: mechanisms and implications. Front Neurosci 2013; 7:144. [PMID: 23964195 PMCID: PMC3740243 DOI: 10.3389/fnins.2013.00144] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/24/2013] [Indexed: 12/20/2022] Open
Abstract
Our early life nutritional environment can influence several aspects of physiology, including our propensity to become obese. There is now evidence to suggest perinatal diet can also independently influence development of our innate immune system. This review will address three not-necessarily-exclusive mechanisms by which perinatal nutrition can program neuroimmune function long-term: by predisposing the individual to obesity, by altering the gut microbiota, and by inducing epigenetic modifications that alter gene transcription throughout life.
Collapse
Affiliation(s)
- Sarah J Spencer
- School of Health Sciences and Health Innovations Research Institute, RMIT University Melbourne, VIC, Australia
| |
Collapse
|
46
|
Chattha KS, Vlasova AN, Kandasamy S, Rajashekara G, Saif LJ. Divergent immunomodulating effects of probiotics on T cell responses to oral attenuated human rotavirus vaccine and virulent human rotavirus infection in a neonatal gnotobiotic piglet disease model. THE JOURNAL OF IMMUNOLOGY 2013; 191:2446-56. [PMID: 23918983 DOI: 10.4049/jimmunol.1300678] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rotaviruses (RVs) are a leading cause of childhood diarrhea. Current oral vaccines are not effective in impoverished countries where the vaccine is needed most. Therefore, alternative affordable strategies are urgently needed. Probiotics can alleviate diarrhea in children and enhance specific systemic and mucosal Ab responses, but the T cell responses are undefined. In this study, we elucidated the T cell and cytokine responses to attenuated human RV (AttHRV) and virulent human RV (HRV) in gnotobiotic pigs colonized with probiotics (Lactobacillus rhamnosus strain GG [LGG] and Bifidobacterium lactis Bb12 [Bb12]), mimicking gut commensals in breastfed infants. Neonatal gnotobiotic pigs are the only animal model susceptible to HRV diarrhea. Probiotic colonized and nonvaccinated (Probiotic) pigs had lower diarrhea and reduced virus shedding postchallenge compared with noncolonized and nonvaccinated pigs (Control). Higher protection in the Probiotic group coincided with higher ileal T regulatory cells (Tregs) before and after challenge, and higher serum TGF-β and lower serum and biliary proinflammatory cytokines postchallenge. Probiotic colonization in vaccinated pigs enhanced innate serum IFN-α, splenic and circulatory IFN-γ-producing T cells, and serum Th1 cytokines, but reduced serum Th2 cytokines compared with noncolonized vaccinated pigs (Vac). Thus, LGG+Bb12 induced systemic Th1 immunostimulatory effects on oral AttHRV vaccine that coincided with lower diarrhea severity and reduced virus shedding postchallenge in Vac+Pro compared with Vac pigs. Previously unreported intestinal CD8 Tregs were induced in vaccinated groups postchallenge. Thus, probiotics LGG+Bb12 exert divergent immunomodulating effects, with enhanced Th1 responses to oral AttHRV vaccine, whereas inducing Treg responses to virulent HRV.
Collapse
Affiliation(s)
- Kuldeep S Chattha
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | | | | | | | | |
Collapse
|
47
|
Diversity of the intestinal microbiota in different patterns of feeding infants by Illumina high-throughput sequencing. World J Microbiol Biotechnol 2013; 29:2365-72. [PMID: 23793940 DOI: 10.1007/s11274-013-1404-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/17/2013] [Indexed: 12/26/2022]
Abstract
The gastrointestinal microbiota plays a crucial role in the health and disease of the host through its impact on nutrition. Gut microbial composition is related to different diets, but an association of microbiota with different diets in infant has not yet been shown. In this work, we compared the fecal microbiota of breast-fed (BF) and formula-fed infants (FF). By using Illumina high-throughput sequencing and biochemical analyses, we found differences in gut microbiota between the two groups. BF infants showed a significant enrichment of Actinobacteria and Firmicutes and depletion of Proteobacteria (P < 0.05), the abundance of Bacteroidetes in the two groups was very low (P > 0.05). Enterobacteriaceae (Proteobacteria) were the dominant bacteria in FF infant fecal microbiota, and Veillonellaceae (Firmicutes) and Enterobacteriaceae (Proteobacteria) were the dominant bacteria in the BF infant fecal microbiota. The number of genera (percentage of sequences >0.1 %) in BF and FF infants was 17 and 15 respectively, and Streptococcus was the dominant bacterial genus in both groups.
Collapse
|
48
|
Lee JW, Lee JH, Sung SH, Lee SJ. Preventive effects of Lactobacillus mixture on experimental E. coli urinary tract infection in infant rats. Yonsei Med J 2013; 54:489-93. [PMID: 23364986 PMCID: PMC3575986 DOI: 10.3349/ymj.2013.54.2.489] [Citation(s) in RCA: 5] [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] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Urinary tract infection (UTI) is an ascending infection of fecal uropathogens, urogenital lactobacilli are suggested to play a role in the prevention of UTI. This study was to investigate whether lactobacillus mixture (LM) could prevent the experimental infantile UTI. MATERIALS AND METHODS The LM were composed of three lactobacillus strains (L. gasseri, L. rhamnosus, and L. reuteri). Mother rats were grouped as lactobacillus (LB) group I (LB I, n=22), II (LB II, n=24) and control (n=20). LB I and LB II were fed with LM (1 mL/day) and control with phosphate-buffered saline (PBS) from late pregnancy through lactation. All newborn rats were breast-fed and their urine and stool were collected at the end of the 3rd week to compare lactobacillus colony. Then, infant rats from LB II were treated with intravesical instillation of LM. Infant rats from LB I and control were instilled with PBS. Twenty-four hours later, experimental UTI was introduced by intravesical instillation of standard E. coli strain. After 72 hours later, the infant rats were sacrificed for histologic examination. RESULTS Lactobacilli colonies in urine and stool were not statistically different among the three groups. The incidence of pyelonephritis in the LB II was 16.7% (4/24), LB I 72.7% (16.22) and control 75.0% (15/20) (p=0.015). The incidence of cystitis was not significantly different among the three groups. CONCLUSION The intravesically instilled LM significantly prevented experimental pyelonephritis in infant rats, however, LM administered orally to the pregnant and lactating mother rats did not.
Collapse
Affiliation(s)
- Jung Won Lee
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University, Seoul, Korea
| | - Jee Hyun Lee
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University, Seoul, Korea
| | - Sun Hee Sung
- Department of Pathology, Ewha Womans University School of Medicine, Seoul, Korea
| | - Seung Joo Lee
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Chattha KS, Vlasova AN, Kandasamy S, Esseili MA, Siegismund C, Rajashekara G, Saif LJ. Probiotics and colostrum/milk differentially affect neonatal humoral immune responses to oral rotavirus vaccine. Vaccine 2013; 31:1916-23. [PMID: 23453730 DOI: 10.1016/j.vaccine.2013.02.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/05/2013] [Accepted: 02/08/2013] [Indexed: 02/04/2023]
Abstract
Breast milk (colostrum [col]/milk) components and gut commensals play important roles in neonatal immune maturation, establishment of gut homeostasis and immune responses to enteric pathogens and oral vaccines. We investigated the impact of colonization by probiotics, Lactobacillus rhamnosus GG (LGG) and Bifidobacterium lactis Bb12 (Bb12) with/without col/milk (mimicking breast/formula fed infants) on B lymphocyte responses to an attenuated (Att) human rotavirus (HRV) Wa strain vaccine in a neonatal gnotobiotic pig model. Col/milk did not affect probiotic colonization in AttHRV vaccinated pigs. However, unvaccinated pigs fed col/milk shed higher numbers of probiotic bacteria in feces than non-col/milk fed colonized controls. In AttHRV vaccinated pigs, col/milk feeding with probiotic treatment resulted in higher mean serum IgA HRV antibody titers and intestinal IgA antibody secreting cell (ASC) numbers compared to col/milk fed, non-colonized vaccinated pigs. In vaccinated pigs without col/milk, probiotic colonization did not affect IgA HRV antibody titers, but serum IgG HRV antibody titers and gut IgG ASC numbers were lower, suggesting that certain probiotics differentially impact HRV vaccine responses. Our findings suggest that col/milk components (soluble mediators) affect initial probiotic colonization, and together, they modulate neonatal antibody responses to oral AttHRV vaccine in complex ways.
Collapse
Affiliation(s)
- Kuldeep S Chattha
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691-4096, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Matamoros S, Gras-Leguen C, Le Vacon F, Potel G, de La Cochetiere MF. Development of intestinal microbiota in infants and its impact on health. Trends Microbiol 2013; 21:167-73. [PMID: 23332725 DOI: 10.1016/j.tim.2012.12.001] [Citation(s) in RCA: 368] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 12/20/2022]
Abstract
Throughout the human lifetime, the intestinal microbiota performs vital functions, such as barrier function, metabolic reactions, trophic effects, and maturation of the host's innate and adaptive immune responses. Development of the intestinal microbiota in infants is characterized by rapid and large changes in microbial abundance, diversity, and composition. These changes are influenced by medical, cultural, and environmental factors such as mode of delivery, diet, familial environment, diseases, and therapies used. Thus, it is nearly impossible to define a universal standard for intestinal colonization and development of the intestinal microbiota. This review discusses recent data on the early colonization of the gut by microbial species, development of the intestinal microbiota, and its impact on health.
Collapse
Affiliation(s)
- Sebastien Matamoros
- Institut des Nutraceutiques et des Aliments Fonctionnels (INAF), Université Laval, 2440, Boul. Hochelaga, Quebec G1V 0A6, Canada
| | | | | | | | | |
Collapse
|