1
|
Guamán LP, Carrera-Pacheco SE, Zúñiga-Miranda J, Teran E, Erazo C, Barba-Ostria C. The Impact of Bioactive Molecules from Probiotics on Child Health: A Comprehensive Review. Nutrients 2024; 16:3706. [PMID: 39519539 PMCID: PMC11547800 DOI: 10.3390/nu16213706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background: This review investigates the impact of bioactive molecules produced by probiotics on child health, focusing on their roles in modulating gut microbiota, enhancing immune function, and supporting overall development. Key metabolites, including short-chain fatty acids (SCFAs), bacteriocins, exopolysaccharides (EPSs), vitamins, and gamma-aminobutyric acid (GABA), are highlighted for their ability to maintain gut health, regulate inflammation, and support neurodevelopment. Objectives: The aim of this review is to examine the mechanisms of action and clinical evidence supporting the use of probiotics and postbiotics in pediatric healthcare, with a focus on promoting optimal growth, development, and overall health in children. Methods: The review synthesizes findings from clinical studies that investigate the effects of probiotics and their metabolites on pediatric health. The focus is on specific probiotics and their ability to influence gut health, immune responses, and developmental outcomes. Results: Clinical studies demonstrate that specific probiotics and their metabolites can reduce gastrointestinal disorders, enhance immune responses, and decrease the incidence of allergies and respiratory infections in pediatric populations. Additionally, postbiotics-bioactive compounds from probiotic fermentation-offer promising benefits, such as improved gut barrier function, reduced inflammation, and enhanced nutrient absorption, while presenting fewer safety concerns compared to live probiotics. Conclusions: By examining the mechanisms of action and clinical evidence, this review underscores the potential of integrating probiotics and postbiotics into pediatric healthcare strategies to promote optimal growth, development, and overall health in children.
Collapse
Affiliation(s)
- Linda P. Guamán
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Saskya E. Carrera-Pacheco
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Johana Zúñiga-Miranda
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Cesar Erazo
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Carlos Barba-Ostria
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador
| |
Collapse
|
2
|
Babina K, Salikhova D, Polyakova M, Zaytsev A, Egiazaryan A, Novozhilova N. Knowledge and Attitude towards Probiotics among Dental Students and Teachers: A Cross-Sectional Survey. Dent J (Basel) 2023; 11:dj11050119. [PMID: 37232770 DOI: 10.3390/dj11050119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/24/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
This cross-sectional survey assessed the knowledge of and the attitude towards probiotics of dental students and academics at Sechenov University, Moscow, Russia. Our questionnaire consisted of 15 questions divided into 3 sections: respondents' sociodemographic data, knowledge on probiotics, and attitude towards probiotics. The data were analyzed using the Mann-Whitney U test, Fisher's exact test, and Spearman's rank correlation coefficient. Out of the 658 questionnaires distributed, a total of 239 questionnaires were completed by the undergraduates, yielding a response rate of 39.6%, and 54 by the teaching staff (response rate = 100%). Most students (53.6%) and teachers (55.5%) had a fair knowledge of probiotics (p = 0.3135). A vast majority of dental students (97.9%) and all teachers had a positive attitude towards probiotics, with higher mean scores among academics (p < 0.001). A positive weak correlation was found between knowledge and attitude (Spearman r = 0.17, p = 0.0027). The results obtained reveal the need for more evidence-based educational trainings for university teachers and a course on probiotics to be included in the curriculum for dental students.
Collapse
Affiliation(s)
- Ksenia Babina
- Department of Therapeutic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Dilara Salikhova
- Department of Therapeutic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Maria Polyakova
- Department of Therapeutic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Alexandr Zaytsev
- Institute of Linguistics and Intercultural Communication, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Anna Egiazaryan
- Department of Therapeutic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Nina Novozhilova
- Department of Therapeutic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
3
|
Effects of Taro (Colocasia esculenta) Water-Soluble Non-Starch Polysaccharide, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium infantis, and Their Synbiotic Mixtures on Pro-Inflammatory Cytokine Interleukin-8 Production. Nutrients 2022; 14:nu14102128. [PMID: 35631269 PMCID: PMC9147535 DOI: 10.3390/nu14102128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
In the past decades, the regulation of pro-inflammatory cytokine production, including interleukin-8 (IL-8), has been the goal of many targeted therapeutic interventions for Necrotising enterocolitis (NEC), a gastrointestinal disease commonly associated with a very low birth weight in preterm infants. In this study, the ability to regulate the production of IL-8 of the water-soluble non-starch polysaccharide (WS-NSP) from taro corm (Tc-WS-NSP) extracted using a conventional (CE) or improved conventional (ICE) extraction method, of the probiotics Lactobacillus acidophilus, Bifidobacterium breve, and Bifidobacterium infantis, and their synbiotic mixtures were evaluated. The TNF-α stimulated HT-29 cells were incubated with undigested or digested Tc-WS-NSPs (CE or ICE), probiotics, and their synbiotic mixtures with Klebsiella oxytoca, an NEC-positive-associated pathogen. Overall, the synbiotic mixtures of digested Tc-WS-NSP-ICE and high bacterial concentrations of L. acidophilus (5.57 × 109), B. breve (2.7 × 108 CFU/mL), and B. infantis (1.53 × 108) demonstrated higher (42.0%, 45.0%, 43.1%, respectively) ability to downregulate IL-8 compared to the sole use of Tc-WS-NSPs (24.5%), or the probiotics L. acidophilus (32.3%), B. breve (37.8%), or B. infantis (33.1%). The ability demonstrated by the Tc-WS-NSPs, the probiotics, and their synbiotics mixtures to downregulate IL-8 production in the presence of an NEC-positive-associated pathogen may be useful in the development of novel prophylactic agents against NEC.
Collapse
|
4
|
Bungau SG, Tit DM, Vesa CM, Abid A, Szilagyi DV, Radu AF, Bungau AF, Tarce AG, Behl T, Stoicescu M, Brisc CM, Gitea D, Nechifor AC, Endres L. Non-conventional therapeutical approaches to acne vulgaris related to its association with metabolic disorders. Eur J Pharmacol 2022; 923:174936. [PMID: 35378101 DOI: 10.1016/j.ejphar.2022.174936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/12/2022] [Accepted: 03/30/2022] [Indexed: 12/31/2022]
Abstract
The ever-increasing frequency of metabolic syndrome (MetS) is still a major challenge of the public health care system, worldwide. In recent years, researchers have been drawn to the uncommon (at first look) link between skin illnesses and MetS. Because of the pro-inflammatory mechanisms and insulin resistance (IR) that are upregulated in metabolic syndrome, many skin disorders are correlated to metabolic dysfunctions, including acne vulgaris. A comprehensive understanding of the link between MetS and acne vulgaris may contribute to the development of new treatment strategies. The current review focuses on dietary and therapeutic interventions and assesses the effect of various approaches such as improving diet by avoiding certain food products (i.e., milk and chocolate) or increasing the intake of others (i.e., food products rich in omega-3 fatty acids), metformin administration, therapy with plant extracts, plant essential oils, and probiotic supplementation on the improvement of certain acne vulgaris severity parameters. These therapeutic approaches, when combined with allopathic treatment, can improve the patients' quality of life.
Collapse
Affiliation(s)
- Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania; Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
| | - Delia Mirela Tit
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania; Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | - Areha Abid
- Department of Food and Bioproduct Science, College of Agriculture and Bioresources, University of Saskatchewan, Canada.
| | - Denisa-Viola Szilagyi
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087, Oradea, Romania.
| | - Alexa Florina Bungau
- Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | | | - Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India.
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | - Cristina Mihaela Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| | - Daniela Gitea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028, Oradea, Romania.
| | - Aurelia Cristina Nechifor
- Analytical Chemistry and Environmental Engineering Department, University Politehnica of Bucharest, 011061, Bucharest, Romania.
| | - Laura Endres
- Department of Psycho-neurosciences and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073, Oradea, Romania.
| |
Collapse
|
5
|
Sharif S, Heath PT, Oddie SJ, McGuire W. Synbiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2022; 3:CD014067. [PMID: 35230697 PMCID: PMC8887627 DOI: 10.1002/14651858.cd014067.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Intestinal dysbiosis may contribute to the pathogenesis of necrotising enterocolitis (NEC) in very preterm or very low birth weight (VLBW) infants. Dietary supplementation with synbiotics (probiotic micro-organisms combined with prebiotic oligosaccharides) to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of NEC and associated mortality and morbidity. OBJECTIVES To assess the effect of enteral supplementation with synbiotics (versus placebo or no treatment, or versus probiotics or prebiotics alone) for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, the Maternity and Infant Care database and CINAHL, from earliest records to 17 June 2021. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing prophylactic synbiotics supplementation with placebo or no synbiotics in very preterm (< 32 weeks' gestation) or very low birth weight (< 1500 g) infants. DATA COLLECTION AND ANALYSIS Two review authors separately performed the screening and selection process, evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference, with associated 95% confidence intervals (CIs). We used the GRADE approach to assess the level of certainty for effects on NEC, all-cause mortality, late-onset invasive infection, and neurodevelopmental impairment. MAIN RESULTS We included six trials in which a total of 925 infants participated. Most trials were small (median sample size 200). Lack of clarity on methods used to conceal allocation and mask caregivers or investigators were potential sources of bias in four of the trials. The studied synbiotics preparations contained lactobacilli or bifidobacteria (or both) combined with fructo- or galacto-oligosaccharides (or both). Meta-analyses suggested that synbiotics may reduce the risk of NEC (RR 0.18, 95% CI 0.09 to 0.40; RD 70 fewer per 1000, 95% CI 100 fewer to 40 fewer; number needed to treat for an additional beneficial outcome (NNTB) 14, 95% CI 10 to 25; six trials (907 infants); low certainty evidence); and all-cause mortality prior to hospital discharge (RR 0.53, 95% CI 0.33 to 0.85; RD 50 fewer per 1000, 95% CI 120 fewer to 100 fewer; NNTB 20, 95% CI 8 to 100; six trials (925 infants); low-certainty evidence). Synbiotics may have little or no effect on late-onset invasive infection, but the evidence is very uncertain (RR 0.84, 95% CI 0.58 to 1.21; RD 20 fewer per 1000, 95% CI 70 fewer to 30 more; five trials (707 infants); very low-certainty evidence). None of the trials assessed neurodevelopmental outcomes. In the absence of high levels of heterogeneity, we did not undertake any subgroup analysis (including the type of feeding). AUTHORS' CONCLUSIONS The available trial data provide only low-certainty evidence about the effects of synbiotics on the risk of NEC and associated morbidity and mortality for very preterm or very low birth weight infants. Our confidence in the effect estimates is limited; the true effects may be substantially different from these estimates. Large, high-quality trials would be needed to provide evidence of sufficient validity and applicability to inform policy and practice.
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute, St. George's, University of London, London, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
6
|
Hascoët JM, Chevallier M, Gire C, Brat R, Rozé JC, Norbert K, Chen Y, Hartweg M, Billeaud C. Use of a Liquid Supplement Containing 2 Human Milk Oligosaccharides: The First Double-Blind, Randomized, Controlled Trial in Pre-term Infants. Front Pediatr 2022; 10:858380. [PMID: 35601412 PMCID: PMC9119431 DOI: 10.3389/fped.2022.858380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022] Open
Abstract
There is growing evidence supporting the benefit of human milk oligosaccharides (HMOs) on reducing risk of illnesses and improving immune function in newborn infants, but evidence in pre-term infants is lacking. This randomized, double-blind, placebo-controlled trial (NCT03607942) of pre-term infants evaluated the effects of HMO supplementation on feeding tolerance, growth, and safety in 7 neonatal units in France. Pre-term infants (27-33 weeks' gestation, birth weight <1,700 g) were randomized early after birth to receive HMO supplement (n = 43) [2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT) in a 10:1 ratio (0.374 g/kg body weight/day)] or an isocaloric placebo (n = 43) consisting of only glucose (0.140 g/kg/day) until discharge from the neonatal unit. Anthropometric z-scores were calculated using Fenton growth standards. Primary outcome was feeding tolerance, measured by non-inferiority (NI) in days to reach full enteral feeding (FEF) from birth in HMO vs. placebo group (NI margin = 4+ days). Mean number of days on intervention prior to FEF was 8.9 and 10.3 days in HMO and placebo, respectively. Non-inferiority in time to reach FEF in HMO (vs. placebo) was achieved [LS mean difference (95% CI) = -2.16 (-5.33, 1.00); upper bound of 95% CI < NI margin] in full analysis set and similar for per protocol. Adjusted mean time to reach FEF from birth was 2 days shorter in HMO (12.2) vs. placebo (14.3), although not statistically significant (p = 0.177). There was no difference in weight-for-age z-scores between groups throughout the FEF period until discharge. Length-for-age z-scores were higher in HMO at FEF day 14 [0.29 (0.02, 0.56), p = 0.037] and 21 [0.31 (0.02, 0.61), p = 0.037]. Head circumference-for-age z-score was higher in HMO vs. placebo at discharge [0.42 (0.12, 0.71), p = 0.007]. Occurrence of adverse events (AEs) was similar in both groups and relatively common in this population, whereas 2.3 and 14.3%, respectively, experienced investigator-confirmed, related AEs. HMO supplementation is safe and well-tolerated in pre-term infants. After 9 days of supplementation, the HMO group reached FEF 2 days earlier vs. placebo, although the difference was not statistically significant. In addition, HMO supplementation supports early postnatal growth, which may have a positive impact on long-term growth and developmental outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Christophe Rozé
- Hôpital Femme Enfant Adolescent Néonatologie et Réanimation Pédiatrique, Nantes, France
| | | | - Yipu Chen
- Nestlé Product Technology Center-Nutrition, Vevey, Switzerland
| | | | | |
Collapse
|
7
|
Lee Y, Seo H, Kim S, Rahim MDA, Yoon Y, Jung J, Lee S, Beom Ryu C, Song HY. Activity of Lactobacillus crispatus isolated from vaginal microbiota against Mycobacterium tuberculosis. J Microbiol 2021; 59:1019-1030. [PMID: 34724180 DOI: 10.1007/s12275-021-1332-0] [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: 06/23/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
Tuberculosis, an infectious disease, is caused by Mycobacterium tuberculosis. It remains a significant public health issue around the globe, causing about 1.8 million deaths every year. Drug-resistant M. tuberculosis, including multi-drug-resistant (MDR), extremely-drug-resistant (XDR), and totally drug-resistant (TDR) M. tuberculosis, continues to be a threat to public health. In the case of antibiotic-resistant tuberculosis, the treatment effect of conventional antibiotics is low. Side effects caused by high doses over a long period are causing severe problems. To overcome these problems, there is an urgent need to develop a new anti-tuberculosis drug that is different from the existing compound-based antibiotics. Probiotics are defined as live microorganisms conferring health benefits. They can be potential therapeutic agents in this context as the effectiveness of probiotics against different infectious diseases has been well established. Here, we report that Lactobacillus crispatus PMC201 shows a promising effect on tuberculosis isolated from vaginal fluids of healthy Korean women. Lactobacillus crispatus PMC201 reduced M. tuberculosis H37Rv under co-culture conditions in broth and reduced M. tuberculosis H37Rv and XDR M. tuberculosis in macrophages. Lactobacillus crispatus PMC201 was not toxic to a guinea pig model and did not induce dysbiosis in a human intestinal microbial ecosystem simulator. Taken together, these results indicate that L. crispatus PMC201 can be a promising alternative drug candidate in the current tuberculosis drug regime. Further study is warranted to assess the in vivo efficacy and confirm the mode of action of L. crispatus PMC201.
Collapse
Affiliation(s)
- Youngkyoung Lee
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Hoonhee Seo
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sukyung Kim
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - M D Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Youjin Yoon
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Jehee Jung
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Saebim Lee
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Chang Beom Ryu
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, 14584, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
- Probiotics Microbiome Convergence Center, Soonchunhyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
8
|
The Interplay between the Gut Microbiome and the Immune System in the Context of Infectious Diseases throughout Life and the Role of Nutrition in Optimizing Treatment Strategies. Nutrients 2021; 13:nu13030886. [PMID: 33803407 PMCID: PMC8001875 DOI: 10.3390/nu13030886] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
Infectious diseases and infections remain a leading cause of death in low-income countries and a major risk to vulnerable groups, such as infants and the elderly. The immune system plays a crucial role in the susceptibility, persistence, and clearance of these infections. With 70–80% of immune cells being present in the gut, there is an intricate interplay between the intestinal microbiota, the intestinal epithelial layer, and the local mucosal immune system. In addition to the local mucosal immune responses in the gut, it is increasingly recognized that the gut microbiome also affects systemic immunity. Clinicians are more and more using the increased knowledge about these complex interactions between the immune system, the gut microbiome, and human pathogens. The now well-recognized impact of nutrition on the composition of the gut microbiota and the immune system elucidates the role nutrition can play in improving health. This review describes the mechanisms involved in maintaining the intricate balance between the microbiota, gut health, the local immune response, and systemic immunity, linking this to infectious diseases throughout life, and highlights the impact of nutrition in infectious disease prevention and treatment.
Collapse
|
9
|
Nolan LS, Rimer JM, Good M. The Role of Human Milk Oligosaccharides and Probiotics on the Neonatal Microbiome and Risk of Necrotizing Enterocolitis: A Narrative Review. Nutrients 2020; 12:E3052. [PMID: 33036184 PMCID: PMC7600747 DOI: 10.3390/nu12103052] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Preterm infants are a vulnerable population at risk of intestinal dysbiosis. The newborn microbiome is dominated by Bifidobacterium species, though abnormal microbial colonization can occur by exogenous factors such as mode of delivery, formula feeding, and exposure to antibiotics. Therefore, preterm infants are predisposed to sepsis and necrotizing enterocolitis (NEC), a fatal gastrointestinal disorder, due to an impaired intestinal barrier, immature immunity, and a dysbiotic gut microbiome. Properties of human milk serve as protection in the prevention of NEC. Human milk oligosaccharides (HMOs) and the microbiome of breast milk are immunomodulatory components that provide intestinal homeostasis through regulation of the microbiome and protection of the intestinal barrier. Enteral probiotic supplements have been trialed to evaluate their impact on establishing intestinal homeostasis. Here, we review the protective role of HMOs, probiotics, and synbiotic combinations in protecting a vulnerable population from the pathogenic features associated with necrotizing enterocolitis.
Collapse
Affiliation(s)
| | | | - Misty Good
- Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.S.N.); (J.M.R.)
| |
Collapse
|
10
|
The Effect of Probiotic Yogurt Containing Lactobacillus Acidophilus LA-5 and Bifidobacterium Lactis BB-12 on Selected Anthropometric Parameters in Obese Individuals on an Energy-Restricted Diet: A Randomized, Controlled Trial. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10175830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Previous studies using probiotics have shown strain-dependent effects on body mass index (BMI), body mass, or fat mass (FM). The aim of this study was to evaluate how the addition of yogurt containing Lactobacillus acidophilus LA-5 and Bifidobacterium animalis subsp. lactis BB-12 strains to a diet plan affects selected anthropometric parameters in obese people on an energy-restricted diet. Fifty-four subjects aged 20–49 (34.52 ± 9.58) years were included in this study. The recruited subjects were assigned to two subgroups: consuming probiotic yogurt along with a hypocaloric diet (GP) (n–27) or the same diet but without an intentional introduction of yogurt (GRD) (n–27) for 12 weeks. Both GP and GRD decreased body weight, BMI, fat mass and visceral fat by 5.59 kg and 4.71 kg, 1.89 and 1.61 kg/m2, 4.80 kg and 4.07 kg, and 0.68 and 0.65 L, respectively, although the obtained differences were not significant. Analysis of GP and GRD results separately at the beginning and end of the intervention showed that fat loss was substantial in both groups (p < 0.05). Consumption of yogurt containing LA-5 and BB-12 does not significantly improve anthropometric parameters in obese patients.
Collapse
|
11
|
Chen K, Xin J, Zhang G, Xie H, Luo L, Yuan S, Bu Y, Yang X, Ge Y, Liu C. A combination of three probiotic strains for treatment of acute diarrhoea in hospitalised children: an open label, randomised controlled trial. Benef Microbes 2020; 11:339-346. [PMID: 32720832 DOI: 10.3920/bm2020.0046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Acute diarrhoea continues to be a leading cause of morbidity, hospitalisation, and mortality worldwide, and probiotics have been proposed as a complementary therapy in the treatment of acute diarrhoea. The goal of this study is to assess the efficacy and safety of three combined probiotic strains, Bifidobacterium lactis Bi-07, Lactobacillus rhamnosus HN001, and Lactobacillus acidophilus NCFM, as an adjunct to rehydration therapy in treatment of acute watery diarrhoea in hospitalised children. Eligible diarrheal children were randomised into intervention group (IG, n=96, conventional treatment for diarrhoea in combination with probiotics) and control group (CG, n=98, conventional treatment for diarrhoea without probiotics). The primary assessments of this study were duration of diarrhoea and hospital stay and improvement in diarrhoea symptoms. Significantly more children in the IG showed improvements in diarrhoea (defined as a decrease of stool frequency to no more than four times per day and an improved stool consistency within 24-48 h after the treatment) than those in the CG (96.9 vs 79.6%, P<0.05). Children supplemented with the mixed strains had a 22.5 h shorter (121.4±13.7 h vs 143.9±19.8 h) mean duration of diarrhoea and 1.2 d shorter hospital stays (5.1±1.2 d vs 6.3±1.4 d) than children only receiving the rehydration therapy (P<0.05). The prevalence of constipation of children in the IG (3.1%) was markedly lower (P<0.05) than that of children in the CG (13.3%) after treatment. In conclusion, the mixture of three probiotic strains given to children aged 1-3 years resulted in shorter durations of diarrhoea and hospitalisation and a higher percentage of improved children.
Collapse
Affiliation(s)
- K Chen
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - J Xin
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - G Zhang
- Department of Pediatric Intensive Care Unit, Chengdu Women's & Children's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China P.R
| | - H Xie
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - L Luo
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - S Yuan
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - Y Bu
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - X Yang
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - Y Ge
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - C Liu
- School of Exercise and Nutritional Sciences, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182-7251, USA
| |
Collapse
|
12
|
Osakunor DNM, Sengeh DM, Mutapi F. Coinfections and comorbidities in African health systems: At the interface of infectious and noninfectious diseases. PLoS Negl Trop Dis 2018; 12:e0006711. [PMID: 30235205 PMCID: PMC6147336 DOI: 10.1371/journal.pntd.0006711] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There is a disease epidemiological transition occurring in Africa, with increasing incidence of noninfectious diseases, superimposed on a health system historically geared more toward the management of communicable diseases. The persistence and sometimes emergence of new pathogens allows for the occurrence of coinfections and comorbidities due to both infectious and noninfectious diseases. There is therefore a need to rethink and restructure African health systems to successfully address this transition. The historical focus of more health resources on infectious diseases requires revision. We hypothesise that the growing burden of noninfectious diseases may be linked directly and indirectly to or further exacerbated by the existence of neglected tropical diseases (NTDs) and other infectious diseases within the population. Herein, we discuss the health burden of coinfections and comorbidities and the challenges to implementing effective and sustainable healthcare in Africa. We also discuss how existing NTD and infectious disease intervention programs in Africa can be leveraged for noninfectious disease intervention. Furthermore, we explore the potential for new technologies-including artificial intelligence and multiplex approaches-for diagnosis and management of chronic diseases for improved health provision in Africa.
Collapse
Affiliation(s)
- Derick Nii Mensah Osakunor
- Centre for Infection, Immunity and Evolution, Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | | | - Francisca Mutapi
- Centre for Infection, Immunity and Evolution, Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
- National Institute for Health Research, Global Health Research Unit Tackling Infections to Benefit Africa, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Eimeria tenella infection perturbs the chicken gut microbiota from the onset of oocyst shedding. Vet Parasitol 2018; 258:30-37. [PMID: 30105975 DOI: 10.1016/j.vetpar.2018.06.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/14/2022]
Abstract
Coccidiosis is a serious threat to the poultry industry, resulting in substantial economic losses worldwide. The effective development of alternative treatments for coccidiosis that does not involve chemotherapy drugs and does not result in antibiotic resistance relies on gaining a clearer understanding of the interaction between host intestinal microbiota and enteric coccidia. Here, we established an Eimeria tenella infection model in chickens and subsequently monitored the changes in the overall intestinal microbiome using 16S rRNA gene sequencing. We found that the gut (i.e. fecal) microbiota of infected chicken differed from that of uninfected naïve animals. Levels of non-pathogenic bacteria, including Lactobacillus and Faecalibacterium declined, whereas those of pathogenic bacteria, including Clostridium, Lysinibacillus, and Escherichia, increased over time in response to E. tenella infection. Similar dynamic changes of the fecal microbiota were observed in both Arbor Acres broilers and White Leghorn chickens, indicating that the perturbation of the microbiota was directly induced by E. tenella infection. Our findings could be used to further elucidate the serious damage to host health caused by coccidia infection, leading to the development of new effective treatment options for coccidiosis.
Collapse
|
14
|
Morozov V, Hansman G, Hanisch FG, Schroten H, Kunz C. Human Milk Oligosaccharides as Promising Antivirals. Mol Nutr Food Res 2018; 62:e1700679. [PMID: 29336526 DOI: 10.1002/mnfr.201700679] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/23/2017] [Indexed: 11/07/2022]
Abstract
Human milk oligosaccharides (HMOs) are diverse unconjugated carbohydrates that are highly abundant in human breast milk. These glycans are investigated in the context of exhibiting multiple functions in infant growth and development. They seem to provide protection against infectious diseases, including a number of poorly manageable viral infections. Although the potential mechanism of the HMO antiviral protection is rather broad, much of the current experimental work has focused on studying of HMO antiadhesive properties. HMOs may mimic structures of viral receptors and block adherence to target cells, thus preventing infection. Still, the potential of HMOs as a source for new antiviral drugs is relatively unexploited. This can be partly attributed to the extreme complexity of the virus-carbohydrate interactions and technical difficulties in HMO isolation, characterization, and manufacturing procedures. Fortunately, we are currently entering a period of major technological advances that have enabled deeper insights into carbohydrate mediated viral entry, rational selection of HMOs as anti-entry inhibitors, and even evaluation of individual synthetic HMO structures. Here, we provide an up-to-date review on glycan binding studies for rotaviruses, noroviruses, influenza viruses, and human immunodeficiency viruses. We also discuss the preventive and therapeutic potential of HMOs as anti-entry inhibitors and address challenges on the route from fundamental studies to clinical trials.
Collapse
Affiliation(s)
- Vasily Morozov
- Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Grant Hansman
- Schaller Research Group at the University of Heidelberg and the DKFZ, Heidelberg, Germany
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Cologne, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases Unit, University Children's Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Clemens Kunz
- Institute of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
15
|
Galacto-Oligosaccharide/Polidextrose Enriched Formula Protects against Respiratory Infections in Infants at High Risk of Atopy: A Randomized Clinical Trial. Nutrients 2018; 10:nu10030286. [PMID: 29494489 PMCID: PMC5872704 DOI: 10.3390/nu10030286] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Early nutrition affects the risk of atopy and infections through modifications of intestinal microbiota. The Prebiotics in the Prevention of Atopy (PIPA) study was a 24-month randomised, double-blind, placebo-controlled trial. It aimed to evaluate the effects of a galacto-oligosaccharide/polydextrose (GOS/PDX)-formula (PF) on atopic dermatitis (AD) and common infections in infants who were born to atopic parents and to investigate the relationship among early nutrition, gut microbiota and clinical outcomes. Methods: A total of 201 and 199 infants were randomized to receive a PF and standard formula (SF), respectively; 140 infants remained on exclusive breastfeeding (BF). Results: The cumulative incidence of AD and its intensity and duration were not statistically different among the three groups. The number of infants with at least one episode of respiratory infection (RI) and the mean number of episodes until 48 weeks of age were significantly lower in the PF group than in the SF group. The number of patients with recurrent RIs and incidence of wheezing lower RIs until 96 weeks were lower in the PF group than the SF group, but similar to the BF group. Bifidobacteria and Clostridium cluster I colonization increased over time in the PF group but decreased in the SF and BF groups. Bifidobacteria had a protective role in RIs, whereas Clostridium cluster I was associated with atopy protection. Conclusion: The early administration of PF protects against RIs and mediates a species-specific modulation of the intestinal microbiota. Trial registration: clinicaltrial.gov Identifier: NCT02116452.
Collapse
|
16
|
Clark AK, Haas KN, Sivamani RK. Edible Plants and Their Influence on the Gut Microbiome and Acne. Int J Mol Sci 2017; 18:ijms18051070. [PMID: 28513546 PMCID: PMC5454980 DOI: 10.3390/ijms18051070] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/28/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022] Open
Abstract
Acne vulgaris affects most people at some point in their lives. Due to unclear etiology, likely with multiple factors, targeted and low-risk treatments have yet to be developed. In this review, we explore the multiple causes of acne and how plant-based foods and supplements can control these. The proposed causative factors include insulin resistance, sex hormone imbalances, inflammation and microbial dysbiosis. There is an emerging body of work on the human gut microbiome and how it mediates feedback between the foods we eat and our bodies. The gut microbiome is also an important mediator of inflammation in the gut and systemically. A low-glycemic load diet, one rich in plant fibers and low in processed foods, has been linked to an improvement in acne, possibly through gut changes or attenuation of insulin levels. Though there is much interest in the human microbiome, there is much more unknown, especially along the gut-skin axis. Collectively, the evidence suggests that approaches such as plant-based foods and supplements may be a viable alternative to the current first line standard of care for moderate acne, which typically includes antibiotics. Though patient compliance with major dietary changes is likely much lower than with medications, it is a treatment avenue that warrants further study and development.
Collapse
Affiliation(s)
- Ashley K Clark
- School of Medicine, University of California-Davis, Sacramento, CA 95816, USA.
| | - Kelly N Haas
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA.
| | - Raja K Sivamani
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA.
- Department of Biological Sciences, California State University, Sacramento, CA 95819, USA.
| |
Collapse
|
17
|
Oliveira LFF, Salvador SL, Silva PHF, Furlaneto FAC, Figueiredo L, Casarin R, Ervolino E, Palioto DB, Souza SLS, Taba M, Novaes AB, Messora MR. Benefits of Bifidobacterium animalis subsp. lactis Probiotic in Experimental Periodontitis. J Periodontol 2016; 88:197-208. [PMID: 27660886 DOI: 10.1902/jop.2016.160217] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND This study evaluates effects of topical administration of probiotic bacteria of the genus Bifidobacterium on experimental periodontitis (EP) in rats. METHODS Thirty-two rats were divided into groups C (control; without EP), EP (EP only), C-HN019 (control+probiotic), and EP-HN019 (EP+probiotic). On day 0 of the experiment, animals of groups EP and EP-HN019 received cotton ligatures around mandibular first molars (MFMs). In groups C-HN019 and EP-HN019, 1 mL of suspensions containing Bifidobacterium animalis subsp. lactis (B. lactis) HN019 was topically administered in the subgingival region of MFMs on days 0, 3, and 7. In groups C and EP, topical administrations were performed using a sham suspension (without probiotic). All animals were euthanized at day 14. Gingival tissue, hemimandibles, and oral biofilm were collected. Data were statistically analyzed (P <0.05). RESULTS Group EP presented greater bone porosity, trabecular separation, and connective tissue attachment loss (CTAL) as well as reduced bone volume than all other groups (P <0.05). In group EP-HN019, there were greater proportions of Actinomyces and Streptococcus-like species and lower proportions of Veillonella parvula, Capnocytophaga sputigena, Eikenella corrodens, and Prevotella intermedia-like species than group EP. Group EP-HN019 presented greater expressions of osteoprotegerin and β-defensins than group EP (P <0.05). Group EP presented greater levels of interleukin-1β and receptor activator of nuclear factor-kappa B ligand than group EP-HN019 (P <0.05). CONCLUSION Topical use of B. lactis HN019 promotes a protective effect against alveolar bone loss and CTALs attributable to EP in rats, modifying immunoinflammatory and microbiologic parameters.
Collapse
Affiliation(s)
- Luiz F F Oliveira
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Sérgio L Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo
| | - Pedro H F Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Flávia A C Furlaneto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Luciene Figueiredo
- Department of Periodontology, Dental Research Division, Guarulhos University, São Paulo, Brazil
| | - Renato Casarin
- Department of Prosthodontics and Periodontics, School of Dentistry, Campinas State University, São Paulo, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, Division of Histology, Dental School of Aracatuba, São Paulo State University, São Paulo, Brazil
| | - Daniela B Palioto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Sérgio L S Souza
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Mario Taba
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Arthur B Novaes
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| | - Michel R Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Vandenplas Y. Probiotics and prebiotics in infectious gastroenteritis. Best Pract Res Clin Gastroenterol 2016; 30:49-53. [PMID: 27048896 DOI: 10.1016/j.bpg.2015.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/23/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
Acute gastroenteritis (AGE) is worldwide a common problem in infants and children. While AGE is still an important cause of morbidity and mortality in developing countries, it is mainly a problem with high socioeconomic impact in the rest of the world. Oral rehydration solutions (ORS) and rapid refeeding remain the cornerstone of the management. However, ORS does not decrease the duration of diarrhea. There is evidence that selected strains of probiotics decrease the duration of AGE with 24 h, both in ambulatory care and in hospitalized children, resulting also in a decrease of the duration of hospitalization. Synbiotics are equally effective as probiotics alone, but prebiotics are not effective. Both pro- and prebiotics have limited to no efficacy in the prevention of AGE. The administration of pre- and probiotics is considered to be safe, even in newborns. Only these pre-, pro and synbiotics that have been clinically tested can be recommended.
Collapse
Affiliation(s)
- Yvan Vandenplas
- UZ Brussel, Department of Paediatrics, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
19
|
Pan M, Wan C, Xie Q, Huang R, Tao X, Shah NP, Wei H. Changes in gastric microbiota induced by Helicobacter pylori infection and preventive effects of Lactobacillus plantarum ZDY 2013 against such infection. J Dairy Sci 2015; 99:970-981. [PMID: 26709179 DOI: 10.3168/jds.2015-10510] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/06/2015] [Indexed: 12/24/2022]
Abstract
Helicobacter pylori is a gram-negative pathogen linked to gastric ulcers and stomach cancer. Gastric microbiota might play an essential role in the pathogenesis of these stomach diseases. In this study, we investigated the preventive effect of a probiotic candidate Lactobacillus plantarum ZDY 2013 as a protective agent against the gastric mucosal inflammation and alteration of gastric microbiota induced by H. pylori infection in a mouse model. Prior to infection, mice were pretreated with or without 400 µL of L. plantarum ZDY 2013 at a concentration of 10(9) cfu/mL per mouse. At 6 wk postinfection, gastric mucosal immune response and alteration in gastric microbiota mice were examined by quantitative real-time PCR and high-throughput 16S rRNA gene amplicon sequencing, respectively. The results showed that L. plantarum ZDY 2013 pretreatment prevented increase in inflammatory cytokines (e.g., IL-1β and IFN-γ) and inflammatory cell infiltration in gastric lamina propria induced by H. pylori infection. Weighted UniFrac principal coordinate analysis showed that L. plantarum ZDY 2013 pretreatment prevented the alteration in gastric microbiota post-H. pylori infection. Linear discriminant analysis coupled with effect size identified 22 bacterial taxa (e.g., Pasteurellaceae, Erysipelotrichaceae, Halomonadaceae, Helicobacteraceae, and Spirochaetaceae) that overgrew in the gastric microbiota of H. pylori-infected mice, and most of them belonged to the Proteobacteria phylum. Lactobacillus plantarum ZDY 2013 pretreatment prevented this alteration; only 6 taxa (e.g., Lachnospiraceae, Ruminococcaceae, and Clostridiaceae), mainly from the taxa of Firmicutes and Bacteroidetes, were dominant in the gastric microbiota of the L. plantarum ZDY 2013 pretreated mice. Administration of L. plantarum ZDY 2013 for 3 wk led to increase in several bacterial taxa (e.g., Rikenella, Staphylococcus, Bifidobacterium), although a nonsignificant alteration was found in the gastric microbiota. Overall, this study demonstrated that L. plantarum ZDY 2013 pretreatment played an important role in preventing gastric mucosal inflammation and gastric microbiota alteration induced by H. pylori infection, and the selective modulation in gastric microbiota posed by this intervention suggested that targeting gastric microbiota through oral administration of probiotics might be an alternative strategy to prevent H. pylori infection.
Collapse
Affiliation(s)
- Mingfang Pan
- College of Life Science, Nanchang University, Nanchang 330031, P. R. China; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Cuixiang Wan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Qiong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Renhui Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China
| | - Nagendra P Shah
- College of Life Science, Nanchang University, Nanchang 330031, P. R. China; Food and Nutritional Science, School of Biological Sciences, University of Hong Kong, Hong Kong.
| | - Hua Wei
- College of Life Science, Nanchang University, Nanchang 330031, P. R. China; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, P. R. China.
| |
Collapse
|
20
|
Szilagyi A. Adaptation to Lactose in Lactase Non Persistent People: Effects on Intolerance and the Relationship between Dairy Food Consumption and Evalution of Diseases. Nutrients 2015; 7:6751-79. [PMID: 26287234 PMCID: PMC4555148 DOI: 10.3390/nu7085309] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 02/07/2023] Open
Abstract
Dairy foods contain complex nutrients which interact with the host. Yet, evolution of lactase persistence has divided the human species into those that can or cannot digest lactose in adulthood. Such a ubiquitous trait has differential effects on humanity. The literature is reviewed to explore how the divide affects lactose handling by lactase non persistent persons. There are two basic differences in digesters. Firstly, maldigesters consume less dairy foods, and secondly, excess lactose is digested by colonic microflora. Lactose intolerance in maldigesters may occur with random lactose ingestion. However, lactose intolerance without maldigestion tends to detract from gaining a clear understanding of the mechanisms of symptoms formation and leads to confusion with regards to dairy food consumption. The main consequence of intolerance is withholding dairy foods. However, regular dairy food consumption by lactase non persistent people could lead to colonic adaptation by the microbiome. This process may mimic a prebiotic effect and allows lactase non persistent people to consume more dairy foods enhancing a favorable microbiome. This process then could lead to alterations in outcome of diseases in response to dairy foods in lactose maldigesters. The evidence that lactose is a selective human prebiotic is reviewed and current links between dairy foods and some diseases are discussed within this context. Colonic adaptation has not been adequately studied, especially with modern microbiological techniques.
Collapse
Affiliation(s)
- Andrew Szilagyi
- Division of Gastroenterology, Department of Medicine, Jewish General Hospital, McGill University School of Medicine; 3755, Chemin de la Cote-Ste-Catherine Rd, Rm E110, Montreal H3T 1E2, QC, Canada.
| |
Collapse
|
21
|
Kishino E, Takemura N, Masaki H, Ito T, Nakazawa M. Dietary lactosucrose suppresses influenza A (H1N1) virus infection in mice. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2015; 34:67-76. [PMID: 26594606 PMCID: PMC4654070 DOI: 10.12938/bmfh.2015-005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
This study examined the effects of lactosucrose
(4G-β-D-galactosylsucrose) on influenza A virus infections
in mice. First, the effects of lactosucrose on fermentation in the
cecum and on immune function were investigated. In female BALB/c mice,
lactosucrose supplementation for 6 weeks promoted cecal fermentation
and increased both secretory IgA (SIgA) levels in feces and total IgA
and IgG2a concentrations in serum. Both the percentage of
CD4+ T cells in Peyer’s patches and the cytotoxic
activity of splenic natural killer (NK) cells increased significantly
in response to lactosucrose. Next, we examined the effects of
lactosucrose on low-dose influenza A virus infection in mice. After 2
weeks of dietary supplementation with lactosucrose, the mice were
infected with low-dose influenza A virus. At 7 days post infection, a
comparison with control mice showed that weight loss was suppressed,
as were viral titers in the lungs. In the spleens of lactosucrose-fed
mice, there was an increase in the percentage of NK cells. Lastly,
mice fed lactosucrose were challenged with a lethal dose of influenza
A virus. The survival rate of these mice was significantly higher than
that of mice fed a control diet. These results suggested that
lactosucrose supplementation suppresses influenza A virus infection by
augmenting innate immune responses and enhancing cellular and mucosal
immunity.
Collapse
Affiliation(s)
- Eriko Kishino
- Carbohydrate Research Laboratory, Ensuiko Sugar Refining Co., Ltd., 1-1-1 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Naho Takemura
- Carbohydrate Research Laboratory, Ensuiko Sugar Refining Co., Ltd., 1-1-1 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Hisaharu Masaki
- Carbohydrate Research Laboratory, Ensuiko Sugar Refining Co., Ltd., 1-1-1 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Tetsuya Ito
- Carbohydrate Research Laboratory, Ensuiko Sugar Refining Co., Ltd., 1-1-1 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Masatoshi Nakazawa
- Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
22
|
Influence of daily consumption of synbiotic soy-based product supplemented with okara soybean by-product on risk factors for cardiovascular diseases. Food Res Int 2015. [DOI: 10.1016/j.foodres.2014.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
Abstract
Prebiotics play a role in the development of intestinal flora. When exposed to unabsorbed food, such as prebiotic carbohydrates, intestinal bacteria produce hydrogen. Increases in hydrogen may signify a slower rate of fermentation or digestion. In this blinded, crossover study, infants (n = 13) consumed formula containing either 4 g/L galactooligosaccharide (GOS) or 4 g/L polydextrose (PDX) + GOS, and breath hydrogen was measured. Breath hydrogen was higher in the PDX/GOS group versus GOS alone (mean ± standard error, 25.35 ± 2.87 ppm vs 13.69 ± 2.87 ppm, P = 0.0001). These results indicate that the formula with PDX/GOS may have undergone slower digestion.
Collapse
|
24
|
van den Nieuwboer M, Brummer RJ, Guarner F, Morelli L, Cabana M, Claassen E. Safety of probiotics and synbiotics in children under 18 years of age. Benef Microbes 2015; 6:615-30. [PMID: 25809217 DOI: 10.3920/bm2014.0157] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study aimed to systematically evaluate safety of probiotics and synbiotics in children ageing 0-18 years. This study is the third and final part in a safety trilogy and an update is provided using the most recent available clinical data (2008-2013) by means of the Common Terminology Clinical Adverse Events (CTCAE version 4.0) classification. Safety aspects are represented and related to number of participants per probiotic strain/culture, study duration, dosage, clinical condition and selected afflictions. Analysis of 74 clinical studies indicated that probiotic and/or synbiotic administration in children is safe with regard to the specific evaluated strains, dosages and duration. The population of children include healthy, immune compromised and obese subjects, as well as subjects with intestinal disorders, infections and inflammatory disorders. This study revealed no major safety concerns, as the adverse events (AEs) were unrelated, or not suspected to be related, to the probiotic or synbiotic product. In general the study products were well tolerated. Overall, AEs occurred more frequent in the control arm compared to children receiving probiotics and/or synbiotics. Furthermore, the results indicate inadequate reporting and classification of AEs in the majority of the studies. In addition, generalizability of conclusions are greatly limited by the inconsistent, imprecise and potentially incomplete reporting as well as the variation in probiotic strains, dosages, administration regimes, study populations and reported outcomes.
Collapse
Affiliation(s)
- M van den Nieuwboer
- 1 VU University Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - R J Brummer
- 2 School of Health and Medical Sciences, Örebro University, 701 82 Örebro, Sweden
| | - F Guarner
- 3 Digestive System Research Unit, CIBERehd, University Hospital Vall d'Hebron, 08035 Barcelona, Spain
| | - L Morelli
- 4 Istituto di Microbiologia, Università Cattolica S.C., Via Emilia Parmense 84, 29122 Piacenza, Italy
| | - M Cabana
- 5 University of California San Francisco (UCSF), Departments of Pediatrics, Epidemiology and Biostatistics, 3333 California Street, #245, San Francisco, CA 94118, USA
| | - E Claassen
- 1 VU University Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.,6 Erasmus Medical Center, Department of Viroscience, P.O. Box 2040, 3000 CA Rotterdam, the Netherlands
| |
Collapse
|
25
|
Chiu YH, Lin SL, Tsai JJ, Lin MY. Probiotic actions on diseases: implications for therapeutic treatments. Food Funct 2014; 5:625-34. [PMID: 24549263 DOI: 10.1039/c3fo60600g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The ecology of gut microflora, which colonizes all body surfaces, has long coevolved with its hosts in a complicated fashion. Health benefits conferred by gut microflora include defense against invading pathogens, improvement of nutritional bioavailability, and development of the regional and systemic immune systems. The past decade has witnessed growing interest in the fact that the gut microflora affects the host's energy homeostasis by means of various mechanisms, including supplying nourishment from indigestible compounds, producing small biomolecules responsible for lipid profiles, and participating in the absorption, distribution, metabolism and excretion of nutrition. Much in vitro and in vivo research has indicated that aberrant gut microflora plays an important role in the pathogenesis of a wide spectrum of diseases. This is accomplished by a shift in focus, from laying an emphasis on pharmacotherapy to placing more effort on gut microflora normalization. The objectives of this review include illustrating trends in the clinical application of probiotics on diseases, as well as discussing current methodology limitations on probiotic selection. Furthermore, it is expected to shed light on the nature of probiotics, with the aim of giving greater insight into the implications for clinical use of probiotics in the treatment of diseases.
Collapse
Affiliation(s)
- Yi-Heng Chiu
- Department of Food Science and Biotechnology, National Chung Hsing University, 250 Kuokuang Road, Taichung 40227, Taiwan, ROC.
| | | | | | | |
Collapse
|
26
|
Bridgman SL, Azad MB, Field CJ, Letourneau N, Johnston DW, Kaplan BJ, Kozyrskyj AL. Maternal perspectives on the use of probiotics in infants: a cross-sectional survey. Altern Ther Health Med 2014; 14:366. [PMID: 25267264 PMCID: PMC4193129 DOI: 10.1186/1472-6882-14-366] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/27/2014] [Indexed: 01/09/2023]
Abstract
Background Probiotic products that may modify the intestinal microbiota are becoming increasingly available and known to consumers due to their potential to prevent or treat many pediatric health conditions. As scientific knowledge of the health benefits of probiotics increases, it is important to identify factors that may prevent their successful integration into patient care as well as to ensure effective translation of research findings. The aim of this study was to describe maternal perspectives on probiotics and their use in infants. Methods Mothers with a child aged two years or younger enrolled in the Alberta Pregnancy Outcomes and Nutrition (APrON) study were invited by email to complete a 29 item self-administered web-based questionnaire. Results A total of 413 mothers of the 1327 contacted completed the questionnaire. The majority (99.3%) of respondents had heard of probiotics and were aware that they contained live bacteria (87.0%); 89.3% had used a product containing probiotics themselves but only 50.8% had given one to their infant. Most mothers indicated they believed that probiotics were beneficial (73.1%) and none thought they were harmful. Over a third of mothers did not feel informed enough to make a decision on whether probiotics were safe to use in infants (36.6%). Conclusions The study demonstrates that awareness and understanding of probiotics is high among mothers in Alberta, Canada. However, there is still uncertainty regarding the benefit of probiotics as well as safety in infants which could be important factors determining therapeutic use in the future. Further studies that demonstrate beneficial effects and safety of probiotics in healthy infants as well as targeted knowledge translation should help to address these potential concerns.
Collapse
|
27
|
Tropical fruit pulps decreased probiotic survival to in vitro gastrointestinal stress in synbiotic soy yoghurt with okara during storage. Lebensm Wiss Technol 2014. [DOI: 10.1016/j.lwt.2013.10.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Luoto R, Ruuskanen O, Waris M, Kalliomäki M, Salminen S, Isolauri E. Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: a randomized, placebo-controlled trial. J Allergy Clin Immunol 2013; 133:405-13. [PMID: 24131826 PMCID: PMC7112326 DOI: 10.1016/j.jaci.2013.08.020] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/15/2013] [Accepted: 08/26/2013] [Indexed: 12/28/2022]
Abstract
Background Simple and safe strategies for the prevention of viral respiratory tract infections (RTIs) are needed. Objective We hypothesized that early prebiotic or probiotic supplementation would reduce the risk of virus-associated RTIs during the first year of life in a cohort of preterm infants. Methods In this randomized, double-blind, placebo-controlled trial (ClinicalTrials.gov no. NCT00167700), 94 preterm infants (gestational age, ≥32 + 0 and ≤36 + 6 weeks; birth weight, >1500 g) treated at Turku University Hospital, Turku, Finland, were allocated to receive oral prebiotics (galacto-oligosaccharide and polydextrose mixture, 1:1), a probiotic (Lactobacillus rhamnosus GG, ATCC 53103), or placebo (microcrystalline cellulose) between days 3 and 60 of life. The primary outcome was the incidence of clinically defined virus-associated RTI episodes confirmed from nasal swabs by using nucleic acid testing. Secondary outcomes were the severity and duration of RTIs. Results A significantly lower incidence of RTIs was detected in infants receiving prebiotics (rate ratio [RR], 0.24; 95% CI, 0.12-0.49; P < .001) or probiotics (RR, 0.50; 95% CI, 0.28-0.90; P = .022) compared with those receiving placebo. Also, the incidence of rhinovirus-induced episodes, which comprised 80% of all RTI episodes, was found to be significantly lower in the prebiotic (RR, 0.31; 95% CI, 0.14-0.66; P = .003) and probiotic (RR, 0.49; 95% CI, 0.24-1.00; P = .051) groups compared with the placebo group. No differences emerged among the study groups in rhinovirus RNA load during infections, duration of rhinovirus RNA shedding, duration or severity of rhinovirus infections, or occurrence of rhinovirus RNA in asymptomatic infants. Conclusions Gut microbiota modification with specific prebiotics and probiotics might offer a novel and cost-effective means to reduce the risk of rhinovirus infections.
Collapse
Affiliation(s)
- Raakel Luoto
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland.
| | - Olli Ruuskanen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Matti Waris
- Department of Virology, University of Turku, Turku, Finland
| | - Marko Kalliomäki
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Erika Isolauri
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
29
|
Wittmann A, Autenrieth IB, Frick JS. Plasmacytoid dendritic cells are crucial in Bifidobacterium adolescentis-mediated inhibition of Yersinia enterocolitica infection. PLoS One 2013; 8:e71338. [PMID: 23977019 PMCID: PMC3748105 DOI: 10.1371/journal.pone.0071338] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/03/2013] [Indexed: 12/23/2022] Open
Abstract
In industrialized countries bacterial intestinal infections are commonly caused by enteropathogenic Enterobacteriaceae. The interaction of the microbiota with the host immune system determines the adequacy of an appropriate response against pathogens. In this study we addressed whether the probiotic Bifidobacterium adolescentis is protective during intestinal Yersinia enterocolitica infection. Female C57BL/6 mice were fed with B. adolescentis, infected with Yersinia enterocolitica, or B. adolescentis fed and subsequently infected with Yersinia enterocolitica. B. adolescentis fed and Yersinia infected mice were protected from Yersinia infection as indicated by a significantly reduced weight loss and splenic Yersinia load when compared to Yersinia infected mice. Moreover, protection from infection was associated with increased intestinal plasmacytoid dendritic cell and regulatory T-cell frequencies. Plasmacytoid dendritic cell function was investigated using depletion experiments by injecting B. adolescentis fed, Yersinia infected C57BL/6 mice with anti-mouse PDCA-1 antibody, to deplete plasmacytoid dendritic cells, or respective isotype control. The B. adolescentis-mediated protection from Yersinia dissemination to the spleen was abrogated after plasmacytoid dendritic cell depletion indicating a crucial function for pDC in control of intestinal Yersinia infection. We suggest that feeding of B. adolescentis modulates the intestinal immune system in terms of increased plasmacytoid dendritic cell and regulatory T-cell frequencies, which might account for the B. adolescentis-mediated protection from Yersinia enterocolitica infection.
Collapse
Affiliation(s)
- Alexandra Wittmann
- Institute for Medical Microbiology and Hygiene, University Hospital Tübingen, Tübingen, Germany
- German Centre for Infection Research, University of Tübingen, Tübingen, Germany
| | - Ingo B. Autenrieth
- Institute for Medical Microbiology and Hygiene, University Hospital Tübingen, Tübingen, Germany
- German Centre for Infection Research, University of Tübingen, Tübingen, Germany
| | - Julia-Stefanie Frick
- Institute for Medical Microbiology and Hygiene, University Hospital Tübingen, Tübingen, Germany
- German Centre for Infection Research, University of Tübingen, Tübingen, Germany
- * E-mail:
| |
Collapse
|
30
|
Probiotics and prebiotics in preventing episodes of acute otitis media in high-risk children: a randomized, double-blind, placebo-controlled study. Pediatr Infect Dis J 2013; 32:810-4. [PMID: 23429555 DOI: 10.1097/inf.0b013e31828df4f3] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Several studies have suggested that probiotics (proB) and/or prebiotics (preB) could reduce the burden of infection in infants and toddlers. We aimed to determine whether follow-up formula supplemented with proB and preB could reduce the risk of acute otitis media (AOM). METHODS In this double-blind, placebo-controlled trial from November 2007 to April 2009, 37 pediatricians in France enrolled children 7 to 13 months of age with high risk of AOM who were randomly assigned to receive follow-up formula supplemented with proB (Streptococcus thermophilus NCC 2496, Streptococcus salivarius DSM 13084, Lactobacillus rhamnosus LPR CGMCC 1.3724) and preB (Raftilose/Raftiline) or follow-up formula alone (placebo). During 12 months, the 2 groups were compared for number of AOM episodes diagnosed (primary outcome) and secondary outcomes by the Poisson model (incidence rate ratio [IRR]) or logistic regression (odds ratio; and 95% confidence interval [95% CI]) after adjustment on covariates of interest. RESULTS We enrolled 224 children (112 in each group). All children were vaccinated (4 doses) with the 7-valent pneumococcal conjugate vaccine; demographic characteristics were similar in the 2 groups. In total, 486 AOM episodes were reported, 249 and 237 in the treatment and control groups, respectively. The treatment and control groups did not differ in incidence of AOM (IRR 1.0, 95% CI: 0.8-1.2), lower respiratory tract infections (IRR 0.9, 0.7-1.2) or number of antibiotic treatment courses (IRR = 1.0, 95% CI: 0.8-1.2). Treatment was not associated with recurrent AOM (odds ratio 1.0, 95% CI: 0.5-1.7). With regard to gastrointestinal disorders, both formulas were well tolerated. CONCLUSION The proB and preB included in follow-up formula given to children at 7 to 13 months of age did not reduce the risk of AOM, recurrent AOM, antibiotic use or lower respiratory tract infections at 1 year.
Collapse
|
31
|
Bedani R, Rossi EA, Isay Saad SM. Impact of inulin and okara on Lactobacillus acidophilus La-5 and Bifidobacterium animalis Bb-12 viability in a fermented soy product and probiotic survival under in vitro simulated gastrointestinal conditions. Food Microbiol 2013; 34:382-9. [PMID: 23541206 DOI: 10.1016/j.fm.2013.01.012] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 01/11/2013] [Accepted: 01/21/2013] [Indexed: 12/19/2022]
Abstract
The effect of inulin and/or okara flour on Lactobacillus acidophilus La-5 and Bifidobacterium animalis Bb-12 viability in a fermented soy product (FSP) and on probiotic survival under in vitro simulated gastrointestinal conditions were investigated throughout 28 days of storage at 4 °C. Employing a 2(2) design, four FSP trials were produced from soymilk fermented with ABT-4 culture (La-5, Bb-12, and Streptococcus thermophilus): FSP (control); FSP-I (with inulin, 3 g/100 mL of soymilk); FSP-O (with okara, 5 g/100 mL); FSP-IO (with inulin + okara, ratio 3:5 g/100 mL). Probiotic viabilities ranged from 8 to 9 log cfu/g during the 28 days of storage, and inulin and/or okara flour did not affect the viability of La-5 and Bb-12. Bb-12 resistance to the artificial gastrointestinal juices was higher than for La-5, since the Bb-12 and La-5 populations decreased approximately 0.6 log cfu/g and 3.8 log cfu/g, respectively, throughout storage period. Even though the protective effect of inulin and/or okara flour on probiotic microorganisms was not significant, when compared to a fresh culture, the FSP matrix improved Bb-12 survival on day 1 of storage and may be considered a good vehicle for Bb-12 and could play an important role in probiotic protection against gastrointestinal juices.
Collapse
Affiliation(s)
- Raquel Bedani
- Departamento de Tecnologia Bioquímico-Farmacêutica, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 580, 05508-000 São Paulo, SP, Brazil
| | | | | |
Collapse
|
32
|
Vandenplas Y, De Greef E, Devreker T, Veereman-Wauters G, Hauser B. Probiotics and Prebiotics in Infants and Children. Curr Infect Dis Rep 2013; 15:251-62. [DOI: 10.1007/s11908-013-0334-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|