1
|
Iddrisu I, Monteagudo-Mera A, Poveda C, Shahzad M, Walton GE, Andrews SC. A review of the effect of iron supplementation on the gut microbiota of children in developing countries and the impact of prebiotics. Nutr Res Rev 2025; 38:229-237. [PMID: 38586996 DOI: 10.1017/s0954422424000118] [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] [Indexed: 04/09/2024]
Abstract
Iron is essential for many physiological functions of the body, and it is required for normal growth and development. Iron deficiency (ID) is the most common form of micronutrient malnutrition and is particularly prevalent in infants and young children in developing countries. Iron supplementation is considered the most effective strategy to combat the risk of ID and ID anaemia (IDA) in infants, although iron supplements cause a range of deleterious gut-related problems in malnourished children. The purpose of this review is to assess the available evidence on the effect of iron supplementation on the gut microbiota during childhood ID and to further assess whether prebiotics offer any benefits for iron supplementation. Prebiotics are well known to improve gut-microbial health in children, and recent reports indicate that prebiotics can mitigate the adverse gut-related effects of iron supplementation in children with ID and IDA. Thus, provision of prebiotics alongside iron supplements has the potential for an enhanced strategy for combatting ID and IDA among children in the developing world. However, further understanding is required before the benefit of such combined treatments of ID in nutritionally deprived children across populations can be fully confirmed. Such enhanced understanding is of high relevance in resource-poor countries where ID, poor sanitation and hygiene, alongside inadequate access to good drinking water and poor health systems, are serious public health concerns.
Collapse
Affiliation(s)
- Ishawu Iddrisu
- Rose Ward, Prospect Park Hospital, Berkshire Healthcare NHS Foundation Trust, Reading, RG30 4EJ, UK
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| | - Andrea Monteagudo-Mera
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Muhammed Shahzad
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
- Faculty of Dentistry, Zarqa University, Zarqa, 13110, Jordan
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Simon C Andrews
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| |
Collapse
|
2
|
Reeves KD, Figuereo YF, Weis VG, Hsu FC, Engevik MA, Krigsman A, Walker SJ. Mapping the geographical distribution of the mucosa-associated gut microbiome in GI-symptomatic children with autism spectrum disorder. Am J Physiol Gastrointest Liver Physiol 2024; 327:G217-G234. [PMID: 38887795 PMCID: PMC11637567 DOI: 10.1152/ajpgi.00101.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by cognitive, behavioral, and communication impairments. In the past few years, it has been proposed that alterations in the gut microbiota may contribute to an aberrant communication between the gut and brain in children with ASD. Consistent with this notion, several studies have demonstrated that children with ASD have an altered fecal microbiota compared with typically developing (TD) children. However, it is unclear where along the length of the gastrointestinal (GI) tract these alterations in microbial communities occur. In addition, the variation between specific mucosa-associated communities remains unknown. To address this gap in knowledge of the microbiome associated with ASD, biopsies from the antrum, duodenum, ileum, right colon, and rectum of children with ASD and age- and sex-matched TD children were examined by 16S rRNA sequencing. We observed an overall elevated abundance of Bacillota and Bacteroidota and a decreased abundance of Pseudomonadota in all GI tract regions of both male and female children with ASD compared with TD children. Further analysis at the genera level revealed unique differences in the microbiome in the different regions of the GI tract in children with ASD compared with TD children. We also observed sex-specific differences in the gut microbiota composition in children with ASD. These data indicate that the microbiota of children with ASD is altered in multiple regions of the GI tract and that different anatomic locations have unique alterations in mucosa-associated bacterial genera.NEW & NOTEWORTHY Analysis in stool samples has shown gut microbiota alterations in children with autism spectrum disorder (ASD) compared with typically developing (TD) children. However, it is unclear which segment(s) of the gut exhibit alterations in microbiome composition. In this study, we examined microbiota composition along the gastrointestinal (GI) tract in the stomach, duodenum, ileum, right colon, and rectum. We found site-specific and sex-specific differences in the gut microbiota of children with ASD, compared with controls.
Collapse
Affiliation(s)
- Kimberly D Reeves
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem North Carolina, United States
| | - Yosauri F Figuereo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Victoria G Weis
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Melinda A Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources, Georgetown, Texas, United States
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| |
Collapse
|
3
|
Hardjo J, Selene NB. Stunting and Gut Microbiota: A Literature Review. Pediatr Gastroenterol Hepatol Nutr 2024; 27:137-145. [PMID: 38818278 PMCID: PMC11134181 DOI: 10.5223/pghn.2024.27.3.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 06/01/2024] Open
Abstract
Stunting, a condition characterized by impaired growth and development in children, remains a major public health concern worldwide. Over the past decade, emerging evidence has shed light on the potential role of gut microbiota modulation in stunting. Gut microbiota dysbiosis has been linked to impaired nutrient absorption, chronic inflammation, altered short-chain fatty acid production, and perturbed hormonal and signaling pathways, all of which may hinder optimal growth in children. This review aims to provide a comprehensive analysis of existing research exploring the bidirectional relationship between stunting and the gut microbiota. Although stunting can alter the gut microbial community, microbiota dysbiosis may exacerbate it, forming a vicious cycle that sustains the condition. The need for effective preventive and therapeutic strategies targeting the gut microbiota to combat stunting is also discussed. Nutritional interventions, probiotics, and prebiotics are among the most promising approaches to modulate the gut microbiota and potentially ameliorate stunting outcomes. Ultimately, a better understanding of the gut microbiota-stunting nexus is vital for guiding evidence-based interventions that can improve the growth and development trajectory of children worldwide, making substantial strides toward reducing the burden of stunting in vulnerable populations.
Collapse
Affiliation(s)
- Jessy Hardjo
- Department of Emergency Medicine, General Hospital Ploso, East Java, Indonesia
| | | |
Collapse
|
4
|
Bai GH, Tsai MC, Lin SC, Hsu YH, Chen SY. Unraveling the interplay between norovirus infection, gut microbiota, and novel antiviral approaches: a comprehensive review. Front Microbiol 2023; 14:1212582. [PMID: 37485533 PMCID: PMC10359435 DOI: 10.3389/fmicb.2023.1212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Norovirus infection is a leading cause of acute gastroenteritis worldwide and can also cause harmful chronic infections in individuals with weakened immune systems. The role of the gut microbiota in the interactions between the host and noroviruses has been extensively studied. While most past studies were conducted in vitro or focused on murine noroviruses, recent research has expanded to human noroviruses using in vivo or ex vivo human intestinal enteroids culture studies. The gut microbiota has been observed to have both promoting and inhibiting effects on human noroviruses. Understanding the interaction between noroviruses and the gut microbiota or probiotics is crucial for studying the pathogenesis of norovirus infection and its potential implications, including probiotics and vaccines for infection control. Recently, several clinical trials of probiotics and norovirus vaccines have also been published. Therefore, in this review, we discuss the current understanding and recent updates on the interactions between noroviruses and gut microbiota, including the impact of norovirus on the microbiota profile, pro-viral and antiviral effects of microbiota on norovirus infection, the use of probiotics for treating norovirus infections, and human norovirus vaccine development.
Collapse
Affiliation(s)
- Geng-Hao Bai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Chen Tsai
- Department of General Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Allergy, Asthma and Immunology, Shuang Ho Hospital, New Taipei, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Shuang Ho Hospital, New Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
6
|
Studies on the Mechanism of the Volatile Oils from Caoguo-4 Decoction in Regulating Spleen Deficiency Diarrhea by Adjusting Intestinal Microbiota. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5559151. [PMID: 35126816 PMCID: PMC8813273 DOI: 10.1155/2022/5559151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022]
Abstract
Background The Caoguo-4 decoction, a classical Mongolian medicine formula, is widely used to treat spleen deficiency diarrhea (SDD) in Mongolian for decades. Previously, the Caoguo-4 decoction volatile oil has been confirmed to be effective in ameliorating symptoms of spleen deficiency diarrhea in an animal model. However, the underlying mechanism of the Caoguo-4 decoction volatile oil is yet to be established. The aim of the current study was to investigate the antidiarrheal effects and mechanism of the Caoguo-4 decoction volatile oil. Method Wistar rats were randomly divided into 5 groups of 10 animals including control, model, positive, Caoguo-4 decoction, and Caoguo-4 decoction volatile oil groups (10 rats in each group). All the rats, besides those in the control group, were induced to develop SDD by a bitter-cold purgation method with Xiaochengqi decoction. The antidiarrheal effect of Caoguo-4 decoction volatile oil was evaluated by pathological section, serum D-xylose and AMS content, plasma MTL content, and gut microbiota analysis via 16S rRNA sequencing. Results The results showed that the developed SDD rat model (model group) had decreased food intake, increased weight loss, soft stool, and bad hair color. When compared with the control group, serum was significantly reduced serum D-xylose and AML but increased MTL levels in the model group (p < 0.05). However, after treatment with either the Caoguo-4 decoction (the decoction group) or Smecta (the positive group) or volatile oil from the Caoguo-4 decoction (the volatile oil group), a significant increase in the serum D-xylose levels was observed. Additionally, AML levels significantly increased in the positive and volatile oil groups, and MTL levels significantly decreased in the decoction and volatile oil groups, when compared with the model group (p < 0.05). The pathological changes of the intestinal mucosa showed that the structure of the epithelium in the villi of the small intestine was affected, deformed, and incomplete in the model group when compared with the control group. However, either the decoction group or the volatile oil group recovered the villous morphology. The results of OTU analysis and alpha diversity analysis of intestinal bacteria showed that the intestinal microbiota of the SDD model rats showed an obvious decrease in richness and diversity of intestinal microbiota. But the intervention treatment of decoction and volatile oil could significantly recover the richness and diversity of intestinal microbiota. Conclusion The intestinal microbiota destroyed in SDD modelling could be significantly improved by the Caoguo-4 decoction volatile oils, which provides reference for clinical medication.
Collapse
|
7
|
Iddrisu I, Monteagudo-Mera A, Poveda C, Pyle S, Shahzad M, Andrews S, Walton GE. Malnutrition and Gut Microbiota in Children. Nutrients 2021; 13:nu13082727. [PMID: 34444887 PMCID: PMC8401185 DOI: 10.3390/nu13082727] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Malnutrition continues to threaten the lives of millions across the world, with children being hardest hit. Although inadequate access to food and infectious disease are the primary causes of childhood malnutrition, the gut microbiota may also contribute. This review considers the evidence on the role of diet in modifying the gut microbiota, and how the microbiota impacts childhood malnutrition. It is widely understood that the gut microbiota of children is influenced by diet, which, in turn, can impact child nutritional status. Additionally, diarrhoea, a major contributor to malnutrition, is induced by pathogenic elements of the gut microbiota. Diarrhoea leads to malabsorption of essential nutrients and reduced energy availability resulting in weight loss, which can lead to malnutrition. Alterations in gut microbiota of severe acute malnourished (SAM) children include increased Proteobacteria and decreased Bacteroides levels. Additionally, the gut microbiota of SAM children exhibits lower relative diversity compared with healthy children. Thus, the data indicate a link between gut microbiota and malnutrition in children, suggesting that treatment of childhood malnutrition should include measures that support a healthy gut microbiota. This could be of particular relevance in sub-Saharan Africa and Asia where prevalence of malnutrition remains a major threat to the lives of millions.
Collapse
Affiliation(s)
- Ishawu Iddrisu
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AX, UK; (I.I.); (A.M.-M.); (C.P.)
| | - Andrea Monteagudo-Mera
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AX, UK; (I.I.); (A.M.-M.); (C.P.)
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AX, UK; (I.I.); (A.M.-M.); (C.P.)
| | - Simone Pyle
- Unilever R&D, Colworth Park, Sharnbrook, Bedfordshire MK44 1LQ, UK;
| | - Muhammad Shahzad
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan;
| | - Simon Andrews
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AX, UK;
| | - Gemma Emily Walton
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AX, UK; (I.I.); (A.M.-M.); (C.P.)
- Correspondence:
| |
Collapse
|
8
|
Matrix Effects on the Delivery Efficacy of Bifidobacterium animalis subsp. lactis BB-12 on Fecal Microbiota, Gut Transit Time, and Short-Chain Fatty Acids in Healthy Young Adults. mSphere 2021; 6:e0008421. [PMID: 34232082 PMCID: PMC8386398 DOI: 10.1128/msphere.00084-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Probiotics are consumed in fermented dairy products or as capsules for their putative health benefits. However, little research has been done to evaluate the effects of the delivery matrix on the health benefits of probiotics in humans. To examine the effects of delivering Bifidobacterium animalis subsp. lactis BB-12 (BB-12) (log10 10 ± 0.5 CFU/day) via a yogurt smoothie versus a capsule, we monitored the fecal microbiota, gut transit times (GTTs), and fecal excretion of short-chain fatty acids (SCFAs) in healthy adults. In a randomized, four-period, crossover study performed in a partially blind manner, 36 adults were recruited and randomly assigned to four treatments: control yogurt smoothie (YS), yogurt smoothie with BB-12 added prefermentation (PRE), yogurt smoothie with BB-12 added postfermentation (POST), and capsule containing BB-12 (CAP). Participants’ fecal microbiota was assessed using 16S rRNA sequencing, GTTs via SmartPill, and fecal SCFAs by gas chromatography (GC) before (baseline) and after each intervention. Participants had significantly higher percentage of Streptococcus after consuming YS versus CAP (P = 0.01). Bifidobacterium-specific terminal restriction fragment length polymorphism analysis revealed a significantly higher percentage of B. animalis after consuming PRE and POST compared to baseline, YS, CAP, and final washout (P < 0.0001). The predominant SCFAs were negatively correlated with GTTs. Consumption of BB-12 delivered in a yogurt smoothie or capsule did not significantly alter the composition of the gut microbiota, GTTs, or fecal SCFA concentration of the study cohort. However, daily consumption of BB-12 in yogurt smoothie may result in higher relative abundance of B. animalis in healthy adults. (This trial has been registered at ClinicalTrials.gov under identifier NCT01399996.) IMPORTANCEBifidobacterium animalis subsp. lactis BB-12 is a probiotic strain that has been used worldwide since 1985. It has commonly been delivered in fermented dairy products for perceived benefits associated with gut health and enhanced immune function. In addition to fermented dairy products, many new probiotic-containing alternatives such as probiotic-containing juice, probiotic-containing chocolate, and capsules have been developed. While these products provide more options for people to access probiotics, little research has been done on the effect of delivery matrix (dairy versus nondairy) on their efficacy in humans. In addition, it was unclear how yogurt fermentation may influence the survival of BB-12 in the product or on its performance in vivo. The significance of our study is in simultaneously assessing the effect of BB-12, alone and in different delivery vehicles, on the gut transit time, fecal short-chain fatty acids, and the composition of the gut microbiota of the study cohort.
Collapse
|
9
|
Shaterian N, Abdi F, Ghavidel N, Alidost F. Role of cesarean section in the development of neonatal gut microbiota: A systematic review. Open Med (Wars) 2021; 16:624-639. [PMID: 33869784 PMCID: PMC8035494 DOI: 10.1515/med-2021-0270] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background The delivery mode is one of the factors affecting the type of colonization of the human gut. Gut colonization affects all stages of the human life cycle, and the type of gut microbiome can contribute to immune system function, the development of some diseases, and brain development; and it has a significant impact on a newborn’s growth and development. Methods Terms defined as MeSH keywords were searched by the databases, and web search engines such as PubMed, ClinicalTrials.gov, Embase, Scopus, ProQuest, Web of Science, and Google Scholar were searched between 2010 and 2020. The quality of each study was assessed according to the Newcastle–Ottawa scale, and seven eligible and high-quality studies were analyzed. Finding The abundances of Bacteroides and Bifidobacterium during the first 3 months of life; Lactobacillus and Bacteroides during the second 3 months of life; Bacteroides and Bifidobacterium during the second 6 months of life; and Bacteroides, Enterobacter, and Streptococcus after the first year of life were higher in vaginal delivery-born infants. While infants born by cesarean section (CS) had higher abundances of Clostridium and Lactobacillus during the first 3 months of life, Enterococcus and Clostridium during the second 3 months of life, and Lactobacillus and Staphylococcus after the first year of life. Discussion Delivery mode can affect the type of the human intestinal microbiota. The CS-born babies had lower colonization rates of Bifidobacterium and Bacteroides, but they had higher colonization rates of Clostridium, Lactobacillus, Enterobacter, Enterococcus, and Staphylococcus. Given the effect of microbiota colonization on neonatal health, it is therefore recommended to conduct further studies in order to investigate the effect of the colonization on the delivery mode and on baby’s growth and development. Application to practice The aim of this study was to investigate the role of CS in the development of the neonatal gut microbiota.
Collapse
Affiliation(s)
- Negin Shaterian
- Student Research Committee, Nursing and Midwifery faculty, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abdi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Nooshin Ghavidel
- Social Determinants of Health Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farzane Alidost
- Student Research Committee, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Valeri F, Endres K. How biological sex of the host shapes its gut microbiota. Front Neuroendocrinol 2021; 61:100912. [PMID: 33713673 DOI: 10.1016/j.yfrne.2021.100912] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/10/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
The gut microbiota is a complex system, consisting of a dynamic population of microorganisms, involved in the regulation of the host's homeostasis. A vast number of factors are driving the gut microbiota composition including diet, antibiotics, environment, and lifestyle. However, in the past decade, a growing number of studies also focused on the role of sex in relationship to changes in the gut microbiota composition in animal experiments as well as in human beings. Despite the progress in investigation techniques, still little is known about the mechanism behind the observed sex-related differences. In this review, we summarized current knowledge on the sex-dependent differences of the intestinal commensals and discuss the probable direct impact of sex hormones and more indirect effects such as dietary habits or antibiotics. While we have to conclude limited data on specific developmental stages, a clear role for sexual hormones and most probably for testosterone emerges.
Collapse
Affiliation(s)
- Francesco Valeri
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz 55131, Germany.
| |
Collapse
|
11
|
The Impact of Age and Pathogens Type on the Gut Microbiota in Infants with Diarrhea in Dalian, China. ACTA ACUST UNITED AC 2020; 2020:8837156. [PMID: 33312314 PMCID: PMC7721492 DOI: 10.1155/2020/8837156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/25/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022]
Abstract
Objective Diarrhea in infants is a serious gastrointestinal dysfunction characterized by vomiting and watery bowel movements. Without proper treatment, infants will develop a dangerous electrolyte imbalance. Diarrhea is accompanied by intestinal dysbiosis. This study compared the gut microbiota between healthy infants and diarrheic infants. It also investigated the effects of age and pathogen type on the gut microbiota of infants with diarrhea, providing data for the proper treatment for diarrhea in infants. Materials and Methods DNA was collected from the fecal samples of 42 Chinese infants with diarrhea and 37 healthy infants. The healthy infants and infants with diarrhea were divided into four age groups: 0-120, 120-180, 180-270, and 270-365 days. Using PCR and 16S rRNA high-throughput sequencing, the diarrhea-causing pathogens in these infants were identified and then categorized into four groups: Salmonella infection, Staphylococcus aureus infection, combined Salmonella and Staphylococcus aureus infection, and others (neither Salmonella nor Staphylococcus aureus). Results The species diversity of gut microbiota in diarrheic infants was significantly reduced compared with that in healthy infants. Infants with diarrhea had a lower abundance of Lactobacillus spp. and Bacillus spp. (P < 0.001) and a significant richness of Klebsiella spp. and Enterobacter spp. (P < 0.001). Similar gut microbiota patterns were found in diarrheic infants in all four age groups. However, different pathogenic infections have significant effects on the gut microbiota of diarrheic infants. For instance, the relative abundance of Klebsiella spp. and Streptococcus spp. was significantly increased (P < 0.001) in infants infected with Staphylococcus aureus; meanwhile, the richness of bacteria such as Enterobacter spp. was significantly increased in the Salmonella infection group (P < 0.001). Conclusion The microbiota in infants with diarrhea has changed significantly, characterized by decreased species diversity and abundance of beneficial bacteria and significant increase in the proportion of conditional pathogens. Meanwhile, the gut microbiota of infants with diarrhea at different ages was similar, but different pathogenic infections affect the gut microbiota characteristics. Therefore, early identification of changes in gut microbiota in infants with diarrhea and the adoption of appropriate pathogen type-specific interventions may effectively alleviate the disease and reduce adverse reactions.
Collapse
|
12
|
Xu B, Yan Y, Huang J, Yin B, Pan Y, Ma L. Cortex Phellodendri extract's anti-diarrhea effect in mice related to its modification of gut microbiota. Biomed Pharmacother 2019; 123:109720. [PMID: 31884345 DOI: 10.1016/j.biopha.2019.109720] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/30/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cortex Phellodendri extract (CPE) has been used in China to treat diarrhea whereas the underlying mechanisms remain poorly understood. Given that dysbiosis of gut microbiota is a potential reason for diarrhea, and that oral CPE has a low absorption rate in intestine, we hypothesized that modification of gut microbiota is an important factor in CPE's anti-diarrhea effect. To test this hypothesis, we established a diarrhea model by challenging post-weaning mice with oral Enterotoxigenic-Escherichia coli (ETEC), and then the mice were treated with two doses of CPE (80 mg/kg bodyweight and 160 mg/kg bodyweight) or the vehicle control (phosphate buffered saline). Diarrhea indices, inflammatory factors, morphology of jejunum, short-chain fatty acids (SCFAs), and serum endocrine were determined. Modification of gut microbiota was analyzed using 16S rDNA high-throughput sequencing. The changes in functional profiles of gut microbiota were predicted using software PICRUSt. We then explored the association between CPE-responding bacteria and the symptoms indices with the spearman's rank correlation coefficient and significance test. Compared with diarrheal mice, CPE decreased Gut/Carcass ratio and water content of stool, increased goblet cell density and villus height/crypt depth of jejunum, as well as decreased inflammatory indices (Tumour Necrosis Factor-α, Myeloperoxidase and Interleukin-1α). CPE shifted the gut microbiota significantly by increasing alpha diversity (observed species, ace, Shannon, and Simpson) and restoring the gut microbiota. CPE increased Firmicutes and decreased Bacteroidetes. The reduced genus Prevotella, Acinetobacter, and Morganella were positively associated with the diarrhea indices, whereas increased genus Odoribacter, Rikenella, and Roseburia were negatively associated with the diarrhea indices. The abundance of carbohydrate metabolism-related gene and SCFAs-producing bacteria were increased, which was evidenced by increased butyric acid and total SCFAs concentration in the caecum. Consequently, endocrine peptides glucagon-like peptide-1, epidermal growth factor, and peptide tyrosine tyrosine in serum were elevated. CONCLUSIONS: CPE shows a shift function on the gut microbiota in alleviating the diarrhea of mice in a dose-dependent manner. In addition, the microbial metabolites SCFAs may mediate CPE's anti-diarrhea effect by enhancing endocrine secretion in mice.
Collapse
Affiliation(s)
- Baoyang Xu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yiqin Yan
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Juncheng Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Boqi Yin
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yunxin Pan
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Libao Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China; Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China.
| |
Collapse
|
13
|
Piperata BA, Lee S, Mayta Apaza AC, Cary A, Vilchez S, Oruganti P, Garabed R, Wilson W, Lee J. Characterization of the gut microbiota of Nicaraguan children in a water insecure context. Am J Hum Biol 2019; 32:e23371. [PMID: 31859435 DOI: 10.1002/ajhb.23371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES The gut microbiota varies across human populations. The first years of life are a critical period in its development. While delivery mode and diet contribute to observed variation, the additional contribution of specific environmental factors remains poorly understood. One factor is waterborne enteric pathogen exposure. In this pilot study, we explore the relationship between household water security and the gut microbiota of children. METHODS From Nicaraguan households (n = 39), we collected drinking water samples, as well as fecal samples from children aged one month to 5.99 years (n = 53). We tested water samples for total coliforms (CFU/mL) and the presence of common enteric pathogens. Composition and diversity of the gut microbiota were characterized by 16S rRNA sequencing. Households were classified as having drinking water that was "low" (<29 CFU/mL) or "high" (≥29 CFU/mL) in coliforms. We used permutational analyses of variance and Mann-Whitney U-tests to identify differences in the composition and diversity of the gut microbiota of children living in these two home types. RESULTS Insecure access led households to store drinking water and 85% tested positive for coliforms. High concentrations of Salmonella and Campylobacter were found in water and fecal samples. Controlling for age, the gut microbiota of children from high coliform homes were compositionally different and less diverse than those from low coliform homes. CONCLUSIONS Results indicate that research exploring the ways water insecurity affects human biology should consider the gut microbiome and that investigations of inter-population variation in the gut microbial community of children should consider pathogen exposure and infection.
Collapse
Affiliation(s)
| | - Seungjun Lee
- College of Public Health, Division of Environmental Health Sciences, The Ohio State University, Columbus, Ohio
| | - Alba C Mayta Apaza
- Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| | - Adelaide Cary
- Department of Anthropology, The Ohio State University, Columbus, Ohio
| | - Samuel Vilchez
- Department of Microbiology, National Autonomous University of Nicaragua, León, Nicaragua
| | - Pallavi Oruganti
- College of Veterinary Medicine, Department of Preventative Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Rebecca Garabed
- College of Veterinary Medicine, Department of Preventative Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Warren Wilson
- Department of Anthropology and Archaeology, University of Calgary, Calgary, Canada
| | - Jiyoung Lee
- College of Public Health, Division of Environmental Health Sciences, The Ohio State University, Columbus, Ohio.,Department of Food Science and Technology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
14
|
The Effects of Fecal Donors with Different Feeding Patterns on Diarrhea in a Patient Undergoing Hematopoietic Stem Cell Transplantation. Case Rep Hematol 2019; 2019:4505238. [PMID: 31049232 PMCID: PMC6462343 DOI: 10.1155/2019/4505238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/20/2018] [Accepted: 02/27/2019] [Indexed: 12/14/2022] Open
Abstract
Almost 90% of patients undergoing hematopoietic stem cell transplantation (HSCT) experience diarrheal episodes, which represent a severe, often life-threatening complication for these patients. Although fecal microbiota transplantation (FMT) represents an alternative treatment option for infection-related diarrhea, the application of FMT in HSCT patients is greatly restricted for safety reasons. Furthermore, the therapeutic outcomes of FMT as a diarrhea treatment are somewhat related to the choice of the FMT donor. Here, we comprehensively profiled the dynamic changes in the intestinal microbiota after FMT from two donors with different feeding patterns and the same severely diarrheal recipient undergoing HSCT via a 45-day clinical observation. Importantly, no adverse events attributed to FMT were observed. The stool volume and frequency of the patient were reduced when we used feces from donor #1 (mixed feeding), but these changes were not observed after FMT from donor #2 (exclusive breastfeeding). Interestingly, no obvious differences in overall diversity (Shannon) or richness (Chao1) between the two donors were observed. Additionally, Bifidobacterium accounted for 29.9% and 18.1% of OTUs in the stools of donors #1 and #2, respectively. Lactobacillus accounted for 16.3% and 2.9% of the stools of donors #1 and #2, respectively. Furthermore, through longitudinal monitoring of the patient, we identified 6 OTUs that were particularly sensitive to the different FMT complements. Together, we present a case report suggesting that the overall diversity of the intestinal microbiota may not be the only important element in the selection of an effective FMT donor.
Collapse
|
15
|
Shahramian I, Kalvandi G, Javaherizadeh H, Khalili M, Noori NM, Delaramnasab M, Bazi A. The effects of prebiotic supplementation on weight gain, diarrhoea, constipation, fever and respiratory tract infections in the first year of life. J Paediatr Child Health 2018; 54:875-880. [PMID: 29579345 DOI: 10.1111/jpc.13906] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/28/2018] [Accepted: 02/22/2018] [Indexed: 11/28/2022]
Abstract
AIM This study was conducted to evaluate the effect of prebiotics on some common clinical ailments in healthy term infants. METHODS Sixty healthy-term, breastfed (BF) infants were included. Along with these infants, 120 healthy-term formula-fed infants were randomly assigned to either the prebiotic formula (PF, n = 60) or regular formula (RF, n = 60) groups. Ready-to-use prebiotic-supplemented formula containing galacto-oligosaccharides and polydextrose (ratio 1:1) was used. RESULTS At 2 months of age, PF infants demonstrated significantly higher weight gain than BF and RF. At 6 months of age, bodyweight was significantly higher in the RF group compared to BF and PF groups (P < 0.05). Similar results were seen at 8, 10 and 12 months of age. At 10 months of age, the duration of diarrhoea was significantly shorter in PF-fed compared to the RF (P = 0.03) group. A significant difference was found between PF and RF (P < 0.0001) and BF and RF groups (P = 0.002) for diarrhoea duration. Means of constipation episodes per year were 0.03 ± 0.18, 0.433 ± 0.77 and 0.1 ± 0.30 for the BF, RF and PF groups, respectively, with significant difference found between BF and RF (P = 0.006) and PF and RF (P = 0.02). The means of episodes of respiratory tract infections per year for BF, RF and PF groups were 1 ± 0.69, 1.6 ± 0.88 and 1 ± 0.58, respectively (P = 0.01). CONCLUSION Prebiotic-supplemented and regular formula were similar to breast milk regarding prophylactic effects for diarrhoea, constipation and respiratory tract infections in the first year of life. Prebiotic-supplemented formula may be an appropriate substitution for breast milk when breast milk in unavailable.
Collapse
Affiliation(s)
- Iraj Shahramian
- Pediatric Digestive and Hepatic Research Center, Zabol University of Medical Sciences, Zabol, Iran
| | | | - Hazhir Javaherizadeh
- Department of Pediatric Gastroenterology, Abuzar Children's Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manijeh Khalili
- Children and Adolescent Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Noor Mohammad Noori
- Children and Adolescent Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mojtaba Delaramnasab
- Clinical Research Development Unit, Amir-Al-Momenin Hospital, Zabol University of Medical Sciences, Zabol, Iran
| | - Ali Bazi
- Pediatric Digestive and Hepatic Research Center, Zabol University of Medical Sciences, Zabol, Iran.,Clinical Research Development Unit, Amir-Al-Momenin Hospital, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
16
|
The human gut microbiome of Latin America populations: a landscape to be discovered. Curr Opin Infect Dis 2017; 29:528-37. [PMID: 27479024 DOI: 10.1097/qco.0000000000000300] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The gut microbiome is critical for human health, and its alteration is associated with intestinal, autoimmune and metabolic diseases. Numerous studies have focused on prevention or treatment of dysbiotic microbiome to reduce the risk or effect of these diseases. A key issue is to define the microbiome associated with the state of good health. The purpose of this review is to describe factors influencing the gut microbiome with special emphasis on contributions from Latin America. In addition, we will highlight opportunities for future studies on gut microbiome in Latin America. RECENT FINDINGS A relevant factor influencing gut microbiome composition is geographical location associated with specific genetic, dietary and lifestyle factors. Geographical specificities suggest that a universal 'healthy microbiome' is unlikely. SUMMARY Several research programs, mostly from Europe and North America, are extensively sequencing gut microbiome of healthy people, whereas data from Latin America remain scarce yet slowly increasing. Few studies have shown difference in the composition of gut microbiome between their local populations with that of other industrialized countries (North American populations). Latin America is composed of countries with a myriad of lifestyles, traditions, genetic backgrounds and socioeconomic conditions, which may determine differences in gut microbiome of individuals from different countries. This represents an opportunity to better understand the relationship between these factors and gut microbiome.
Collapse
|
17
|
Chronic Health Consequences of Acute Enteric Infections in the Developing World. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
18
|
Rutayisire E, Huang K, Liu Y, Tao F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants' life: a systematic review. BMC Gastroenterol 2016; 16:86. [PMID: 27475754 PMCID: PMC4967522 DOI: 10.1186/s12876-016-0498-0] [Citation(s) in RCA: 395] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/21/2016] [Indexed: 12/14/2022] Open
Abstract
Background The human gut is the habitat for diverse and dynamic microbial ecosystem. The human microbiota plays a critical role in functions that sustain health and is a positive asset in host defenses. Establishment of the human intestinal microbiota during infancy may be influenced by multiple factors including delivery mode. Present review compiles existing evidences on the effect of delivery mode on the diversity and colonization pattern of infants gut microbiota. Methods Two investigators searched for relevant scientific publications from four databases (Pubmed, Medline, Embase, and Web of Science). The last search was performed on September 21, 2015, using key terms ((delivery mode OR caesarean delivery OR cesarean section OR vaginal delivery) AND (gut microbiota OR gut microbiome OR gut microflora OR intestinal microflora OR microbial diversity) AND (infants OR children)). All included studies described at least two types of gut microbiota in relation to delivery mode (caesarean section vs vaginal delivery) and used fecal samples to detect gut microbiota. Results Seven out of 652 retrieved studies met inclusion criteria, were included in systematic analysis. Caesarean Section (CS) was associated with both lower abundance and diversity of the phyala Actinobacteria and Bacteroidetes, and higher abundance and diversity of the phylum Firmicute from birth to 3 months of life. At the colonization level, Bifidobacterium, and Bacteroides genera seems to be significantly more frequent in vaginally delivered infants compared with CS delivered. These infants were more colonized by the Clostridium, and Lactobacillus genera. From the reports, it is tempting to say that delivery mode has less effect on colonization and diversity of Bifidobacteria, Bacteroides, Clostridium, and Lactobacillus genera from the age of 6 to 12 months of life. Conclusion The diversity and colonization pattern of the gut microbiota were significantly associated to the mode of delivery during the first three months of life, however the observed significant differences disappears after 6 months of infants life. The healthy gut microbiota is considered to promote development and maturation of the immune system while abnormal gut is considered as the major cause of severe gastrointestinal infections during the infancy. Further studies should investigate the diversity and colonization levels of infant gut microbiota in relation to the mode of delivery and its broad impact on infants’ health at each stage of life.
Collapse
Affiliation(s)
- Erigene Rutayisire
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China.,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei City, Anhui Province, People's Republic of China
| | - Yehao Liu
- Department of Public Health Inspection and Quarantine Science, School of Public Health, Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei City, Anhui Province, 230032, People's Republic of China. .,Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei City, Anhui Province, People's Republic of China.
| |
Collapse
|
19
|
da Silva Poló T, Peiró JR, Mendes LCN, Ludwig LF, de Oliveira-Filho EF, Bucardo F, Huynen P, Melin P, Thiry E, Mauroy A. Human norovirus infection in Latin America. J Clin Virol 2016; 78:111-9. [DOI: 10.1016/j.jcv.2016.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/09/2016] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
|
20
|
Jang S, Sun J, Chen P, Lakshman S, Molokin A, Harnly JM, Vinyard BT, Urban JF, Davis CD, Solano-Aguilar G. Flavanol-Enriched Cocoa Powder Alters the Intestinal Microbiota, Tissue and Fluid Metabolite Profiles, and Intestinal Gene Expression in Pigs. J Nutr 2016; 146:673-80. [PMID: 26936136 PMCID: PMC4807644 DOI: 10.3945/jn.115.222968] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/12/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Consumption of cocoa-derived polyphenols has been associated with several health benefits; however, their effects on the intestinal microbiome and related features of host intestinal health are not adequately understood. OBJECTIVE The objective of this study was to determine the effects of eating flavanol-enriched cocoa powder on the composition of the gut microbiota, tissue metabolite profiles, and intestinal immune status. METHODS Male pigs (5 mo old, 28 kg mean body weight) were supplemented with 0, 2.5, 10, or 20 g flavanol-enriched cocoa powder/d for 27 d. Metabolites in serum, urine, the proximal colon contents, liver, and adipose tissue; bacterial abundance in the intestinal contents and feces; and intestinal tissue gene expression of inflammatory markers and Toll-like receptors (TLRs) were then determined. RESULTS O-methyl-epicatechin-glucuronide conjugates dose-dependently increased (P< 0.01) in the urine (35- to 204-fold), serum (6- to 186-fold), and adipose tissue (34- to 1144-fold) of pigs fed cocoa powder. The concentration of 3-hydroxyphenylpropionic acid isomers in urine decreased as the dose of cocoa powder fed to pigs increased (75-85%,P< 0.05). Compared with the unsupplemented pigs, the abundance ofLactobacillusspecies was greater in the feces (7-fold,P= 0.005) and that ofBifidobacteriumspecies was greater in the proximal colon contents (9-fold,P= 0.01) in pigs fed only 20 or 10 g cocoa powder/d, respectively. Moreover, consumption of cocoa powder reducedTLR9gene expression in ileal Peyer's patches (67-80%,P< 0.05) and mesenteric lymph nodes (43-71%,P< 0.05) of pigs fed 2.5-20 g cocoa powder/d compared with pigs not supplemented with cocoa powder. CONCLUSION This study demonstrates that consumption of cocoa powder by pigs can contribute to gut health by enhancing the abundance ofLactobacillusandBifidobacteriumspecies and modulating markers of localized intestinal immunity.
Collapse
Affiliation(s)
| | - Jianghao Sun
- Food Composition and Methods Development Laboratory
| | - Pei Chen
- Food Composition and Methods Development Laboratory
| | | | | | | | - Bryan T Vinyard
- Biometrical Consulting Services, USDA, Northeast Area, Beltsville Human Nutrition Research Center, Beltsville, MD; and
| | | | | | | |
Collapse
|
21
|
Baglatzi L, Gavrili S, Stamouli K, Zachaki S, Favre L, Pecquet S, Benyacoub J, Costalos C. Effect of Infant Formula Containing a Low Dose of the Probiotic Bifidobacterium lactis CNCM I-3446 on Immune and Gut Functions in C-Section Delivered Babies: A Pilot Study. CLINICAL MEDICINE INSIGHTS-PEDIATRICS 2016; 10:11-9. [PMID: 26997881 PMCID: PMC4792197 DOI: 10.4137/cmped.s33096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/04/2015] [Accepted: 11/29/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND In the absence of breast-feeding and its immunomodulatory factors, supplementation of starter infant formula (IF) with probiotics is currently used to support immune functions and gut development. AIM To assess whether immune-related beneficial effects of regular dose (107 CFU/g of powder) of the probiotic Bifidobacterium lactis CNCM I-3446 (hereafter named B. lactis) in starter IF supplementation can be maintained with starter IF containing a low dose (104 CFU/g of powder) of B. lactis. METHOD This trial was designed as a pilot, prospective, double-blind, randomized, single-center clinical trial of two parallel groups (n = 77 infants/group) of C-section delivered infants receiving a starter IF containing either low dose or regular dose of the probiotic B. lactis from birth to six months of age. In addition, a reference group of infants breast-fed for a minimum of four months (n = 44 infants), also born by C-section, were included. All groups were then provided follow-up formula without B. lactis up to 12 months of age. Occurrence of diarrhea, immune and gut maturation, responses to vaccinations, and growth were assessed from birth to 12 months. The effect of low-dose B. lactis formula was compared to regular-dose B. lactis formula, considered as reference for IF with probiotics, and both were further compared to breast-feeding as a physiological reference. RESULTS Data showed that feeding low-dose B. lactis IF provides similar effects as feeding regular-dose B. lactis IF or breast milk. No consistent statistical differences regarding early life protection against gastrointestinal infections, immune and gut maturation, microbiota establishment, and growth were observed between randomized formula-fed groups as well as with the breast-fed reference group. CONCLUSION This pilot study suggests that supplementing C-section born neonates with low-dose B. lactis-containing starter formula may impact immune as well as gut maturation similarly to regular-dose B. lactis, close to the breast-feeding reference.
Collapse
Affiliation(s)
- L Baglatzi
- Neonatal Department, Alexandra Hospital, University of Athens, Greece
| | - S Gavrili
- Neonatal Department, Alexandra Hospital, University of Athens, Greece
| | - K Stamouli
- Neonatal Department, Alexandra Hospital, University of Athens, Greece
| | - S Zachaki
- Neonatal Department, Alexandra Hospital, University of Athens, Greece
| | - L Favre
- Nestlé Research Center, Lausanne, Switzerland
| | - S Pecquet
- Nestlé Nutrition, Vevey, Switzerland
| | - J Benyacoub
- Nestlé Research Center, Lausanne, Switzerland
| | - C Costalos
- Neonatal Department, Alexandra Hospital, University of Athens, Greece
| |
Collapse
|
22
|
Solano-Aguilar G, Molokin A, Botelho C, Fiorino AM, Vinyard B, Li R, Chen C, Urban J, Dawson H, Andreyeva I, Haverkamp M, Hibberd PL. Transcriptomic Profile of Whole Blood Cells from Elderly Subjects Fed Probiotic Bacteria Lactobacillus rhamnosus GG ATCC 53103 (LGG) in a Phase I Open Label Study. PLoS One 2016; 11:e0147426. [PMID: 26859761 PMCID: PMC4747532 DOI: 10.1371/journal.pone.0147426] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 12/31/2015] [Indexed: 02/07/2023] Open
Abstract
We examined gene expression of whole blood cells (WBC) from 11 healthy elderly volunteers participating on a Phase I open label study before and after oral treatment with Lactobacillus rhamnosus GG-ATCC 53103 (LGG)) using RNA-sequencing (RNA-Seq). Elderly patients (65–80 yrs) completed a clinical assessment for health status and had blood drawn for cellular RNA extraction at study admission (Baseline), after 28 days of daily LGG treatment (Day 28) and at the end of the study (Day 56) after LGG treatment had been suspended for 28 days. Treatment compliance was verified by measuring LGG-DNA copy levels detected in host fecal samples. Normalized gene expression levels in WBC RNA were analyzed using a paired design built within three analysis platforms (edgeR, DESeq2 and TSPM) commonly used for gene count data analysis. From the 25,990 transcripts detected, 95 differentially expressed genes (DEGs) were detected in common by all analysis platforms with a nominal significant difference in gene expression at Day 28 following LGG treatment (FDR<0.1; 77 decreased and 18 increased). With a more stringent significance threshold (FDR<0.05), only two genes (FCER2 and LY86), were down-regulated more than 1.5 fold and met the criteria for differential expression across two analysis platforms. The remaining 93 genes were only detected at this threshold level with DESeq2 platform. Data analysis for biological interpretation of DEGs with an absolute fold change of 1.5 revealed down-regulation of overlapping genes involved with Cellular movement, Cell to cell signaling interactions, Immune cell trafficking and Inflammatory response. These data provide evidence for LGG-induced transcriptional modulation in healthy elderly volunteers because pre-treatment transcription levels were restored at 28 days after LGG treatment was stopped. To gain insight into the signaling pathways affected in response to LGG treatment, DEG were mapped using biological pathways and genomic data mining packages to indicate significant biological relevance. Trial Registration: ClinicalTrials.gov NCT01274598
Collapse
Affiliation(s)
- Gloria Solano-Aguilar
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
- * E-mail:
| | - Aleksey Molokin
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Christine Botelho
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Anne-Maria Fiorino
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Bryan Vinyard
- Statistics Group, Northeast Area, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Robert Li
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Celine Chen
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Joseph Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Harry Dawson
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Irina Andreyeva
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Miriam Haverkamp
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Patricia L. Hibberd
- Division of Global Health, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
23
|
Tang Z, Xu L, Shi B, Deng H, Lai X, Liu J, Sun Z. Oral administration of synthetic porcine beta-defensin-2 improves growth performance and cecal microbial flora and down-regulates the expression of intestinal toll-like receptor-4 and inflammatory cytokines in weaned piglets challenged with enterotoxigenic Escherichia coli. Anim Sci J 2015; 87:1258-1266. [PMID: 26707031 DOI: 10.1111/asj.12540] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 08/02/2015] [Accepted: 08/11/2015] [Indexed: 12/26/2022]
Abstract
Synthetic porcine beta-defensin-2 (pBD-2) was tested as an alternative to antimicrobial growth-promoters in pig production. Thirty 21-day weaned piglets were challenged with enterotoxigenic Escherichia coli, and orally dosed with either sterile water (CON), pBD-2 (BD) or neomycin sulphate (NS) twice daily for 21 days. pBD-2 and NS led to higher growth performance, jejunum villus height and increased expression of insulin-like growth factor-I compared with the CON group (P < 0.05). Hemolytic E. coli scores from rectal swabs, and copy numbers of E. coli, Bacteroides fragilis and Streptococcus in the cecal digesta of the BD- or NS-treated piglets were lower than those in the CON group (P < 0.05). Messenger RNA levels of toll-like receptor 4, tumor necrosis factor-α, interleukin (IL)-1β, and IL-8 in the jejunum mucosa of the BD and NS groups were lower than those in the CON group (P < 0.05). Copy numbers of Lactobacilli and Bifidobacteria in the cecal digesta of the BD group were higher than those of the CON and NS groups (P < 0.05). Therefore, pBD-2 has antimicrobial activity in piglets, and it can improve growth performance, reduce inflammatory cytokine expression and affect intestinal morphological indices in the same way as probiotics. © 2015 Japanese Society of Animal Science.
Collapse
Affiliation(s)
- Zhiru Tang
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China.
| | - Ling Xu
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China
| | - Baoshi Shi
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China
| | - Huang Deng
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China
| | - Xin Lai
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China
| | - Jingyan Liu
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China
| | - Zhihong Sun
- Key Laboratory for Bio-feed and Molecular Nutrition, Southwest University, Chongqing, China.
| |
Collapse
|
24
|
Becker-Dreps S, Allali I, Monteagudo A, Vilchez S, Hudgens MG, Rogawski ET, Carroll IM, Zambrana LE, Espinoza F, Azcarate-Peril MA. Gut Microbiome Composition in Young Nicaraguan Children During Diarrhea Episodes and Recovery. Am J Trop Med Hyg 2015; 93:1187-93. [PMID: 26350452 DOI: 10.4269/ajtmh.15-0322] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/23/2015] [Indexed: 12/16/2022] Open
Abstract
Understanding how the gut microbiota is affected by diarrhea episodes may help explain alterations in intestinal function among children in low-income settings. This study examined the composition of the gut microbiome of Nicaraguan children both during diarrhea episodes and while free of diarrhea for at least 2 months. Relative abundances of bacterial taxa, phylogenetic diversity, and species richness were determined by 16S amplicon sequencing and compared between paired diarrhea and recovery samples. A total of 66 stools were provided by 25 children enrolled in a 1-year cohort study of diarrhea etiologies. Children in our cohort had a mean age of 21.9 months; 64% were breast-fed, and 10% had received an antibiotic during the diarrhea episode. Overall, phylogenetic diversity and species richness did not differ significantly between diarrhea and recovery stools. However, of children who had a bacterial enteropathogen detected in any diarrhea stool, none experienced an increase in phylogenetic diversity in recovery, whereas of those in whom no bacterial enteropathogens were detected in their diarrhea stool(s), 59% experienced an increase in phylogenetic diversity in recovery (P = 0.008). This preliminary study suggests that recovery of the gut microbiota after a diarrhea episode may take longer time than previously thought and may be pathogen specific.
Collapse
Affiliation(s)
- Sylvia Becker-Dreps
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Imane Allali
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Andrea Monteagudo
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Samuel Vilchez
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Michael G Hudgens
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Elizabeth T Rogawski
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Ian M Carroll
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Luis Enrique Zambrana
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Felix Espinoza
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - M Andrea Azcarate-Peril
- Department of Family Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Microbiome Core Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Laboratory of Biochemistry and Immunology, Faculty of Sciences, Mohammed V University, Rabat, Morocco; Department of Microbiology and Parasitology, National Autonomous University of Nicaragua, León, Nicaragua; Department of Biostatistics, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Demographic and Health Research, National Autonomous University of Nicaragua, León, Nicaragua; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
25
|
Merenstein DJ, Tan TP, Molokin A, Smith KH, Roberts RF, Shara NM, Mete M, Sanders ME, Solano-Aguilar G. Safety of Bifidobacterium animalis subsp. lactis (B. lactis) strain BB-12-supplemented yogurt in healthy adults on antibiotics: a phase I safety study. Gut Microbes 2015; 6:66-77. [PMID: 25569274 PMCID: PMC4615198 DOI: 10.1080/19490976.2015.1005484] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Probiotics are live microorganisms that, when administered in sufficient doses, provide health benefits on the host. The United States Food and Drug Administration (FDA) requires phase I safety studies for probiotics when the intended use of the product is as a drug. The purpose of the study was to determine the safety of Bifidobacterium animalis subsp lactis (B. lactis) strain BB-12 (BB-12)-supplemented yogurt when consumed by a generally healthy group of adults who were prescribed a 10-day course of antibiotics for a respiratory infection. Secondary aims were to assess the ability of BB-12 to affect the expression of whole blood immune markers associated with cell activation and inflammatory response. A phase I, double-blinded, randomized controlled study was conducted in compliance with FDA guidelines for an Investigational New Drug (IND). Forty participants were randomly assigned to consume 4 ounces of either BB-12 -supplemented yogurt or non-supplemented control yogurt daily for 10 d. The primary outcome was to assess safety and tolerability, assessed by the number of reported adverse events. A total of 165 non-serious adverse events were reported, with no differences between the control and BB-12 groups. When compared to the control group, B lactis fecal levels were modestly higher in the BB-12-supplemented group. In a small subset of patients, changes in whole blood expression of genes associated with regulation and activation of immune cells were detected in the BB-12-supplemented group. BB-12-supplemented yogurt is safe and well tolerated when consumed by healthy adults concurrently taking antibiotics. This study will form the basis for future randomized clinical trials investigating the potential immunomodulatory effects of BB-12-supplemented yogurt in a variety of disease states.
Collapse
Affiliation(s)
- Daniel J Merenstein
- Department of Family Medicine; Georgetown University Medical Center; Washington, DC USA,Correspondence to: Daniel J Merenstein;
| | - Tina P Tan
- Department of Family Medicine; Georgetown University Medical Center; Washington, DC USA
| | - Aleksey Molokin
- United States Department of Agriculture; Agricultural Research Service; Beltsville Human Nutrition Research Center; Diet, Genomics, and Immunology Laboratory; Beltsville, MD USA
| | - Keisha Herbin Smith
- Department of Family Medicine; Georgetown University Medical Center; Washington, DC USA
| | - Robert F Roberts
- Department of Food Science; The Pennsylvania State University; University Park, PA USA
| | - Nawar M Shara
- Department of Biostatistics and Epidemiology; MedStar Health Research Institute; Hyattsville, MD USA
| | - Mihriye Mete
- Department of Biostatistics and Epidemiology; MedStar Health Research Institute; Hyattsville, MD USA
| | | | - Gloria Solano-Aguilar
- United States Department of Agriculture; Agricultural Research Service; Beltsville Human Nutrition Research Center; Diet, Genomics, and Immunology Laboratory; Beltsville, MD USA
| |
Collapse
|