1
|
Noor Z, Hossain B, Mithila NT, Khatun A, Mahmud S, Ferdous A, Akhter B, Alam M, Gilchrist CA, Haque R, Petri WA. Effect of Nitazoxanide and Probiotic Treatment on Bangladeshi Children with Cryptosporidiosis. Am J Trop Med Hyg 2025; 112:733-739. [PMID: 39933176 PMCID: PMC11965742 DOI: 10.4269/ajtmh.23-0914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 11/06/2024] [Indexed: 02/13/2025] Open
Abstract
Cryptosporidium spp. is a cause of diarrhea morbidity and mortality in children under 5 years of age. In addition, asymptomatic infections can have a negative impact on growth and development. In low- and middle-income countries where a greater number of infants may be malnourished, the results of treating cryptosporidiosis with the only Food and Drug Administration-approved drug nitazoxanide (NTZ) have been inconsistent. Malnutrition is both a risk factor for cryptosporidiosis and a consequence of infection with this parasite. Treatment with the probiotic Lactobacillus reuteri DSM 17938 has been shown to assist in nutritional recovery and the restoration of gut health. In this pilot randomized clinical trial, we examined whether combined probiotic and NTZ treatment could result in the reduction in parasitemia and infection-associated growth stunting in undernourished children. Cryptosporidium spp.-positive Bangladeshi children with a weight-for-length Z score between -1 and -3 were randomly assigned to one of three groups. Group 1 (n = 26) received NTZ and Lactobacillus, group 2 (n = 28) received NTZ along with a placebo, and the third control group (n = 10) received standard care. There was no difference in the duration of infection or improvement in child anthropometric measurements in any treatment group compared with control. Therefore, this pilot study does not provide support for treatment with NTZ, Lactobacillus, or the two in combination as an effective means of reducing the duration of Cryptosporidium spp. infection or improving growth in growth-stunted children.
Collapse
Affiliation(s)
- Zannatun Noor
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Biplob Hossain
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Nishad Tasnim Mithila
- Nutrition Research Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Amena Khatun
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sultan Mahmud
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Aleya Ferdous
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Bipasha Akhter
- North South University Genome Research Institute, Dhaka, Bangladesh
| | - Masud Alam
- Department of Molecular Genetics and Microbiology, Vaccine Testing Center, University of Vermont, Burlington, Vermont
| | - Carol A. Gilchrist
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Rashidul Haque
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - William A. Petri
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
2
|
Wall CR, Roy NC, Mullaney JA, McNabb WC, Gasser O, Fraser K, Altermann E, Young W, Cooney J, Lawrence R, Jiang Y, Galland BC, Fu X, Tonkie JN, Mahawar N, Lovell AL. Nourishing the Infant Gut Microbiome to Support Immune Health: Protocol of SUN (Seeding Through Feeding) Randomized Controlled Trial. JMIR Res Protoc 2024; 13:e56772. [PMID: 39222346 PMCID: PMC11406106 DOI: 10.2196/56772] [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: 01/25/2024] [Revised: 05/03/2024] [Accepted: 06/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The introduction of complementary foods during the first year of life influences the diversity of the gut microbiome. How this diversity affects immune development and health is unclear. OBJECTIVE This study evaluates the effect of consuming kūmara or kūmara with added banana powder (resistant starch) compared to a reference control at 4 months post randomization on the prevalence of respiratory tract infections and the development of the gut microbiome. METHODS This study is a double-blind, randomized controlled trial of mothers and their 6-month-old infants (up to n=300) who have not yet started solids. Infants are randomized into one of 3 groups: control arm (C), standard kūmara intervention (K), and a kūmara intervention with added banana powder product (K+) to be consumed daily for 4 months until the infant is approximately 10 months old. Infants are matched for sex using stratified randomization. Data are collected at baseline (prior to commencing solid food) and at 2 and 4 months after commencing solid food (at around 8 and 10 months of age). Data and samples collected at each timepoint include weight and length, intervention adherence (months 2 and 4), illness and medication history, dietary intake (months 2 and 4), sleep (diary and actigraphy), maternal dietary intake, breast milk, feces (baseline and 4 months), and blood samples (baseline and 4 months). RESULTS The trial was approved by the Health and Disability Ethics Committee of the Ministry of Health, New Zealand (reference 20/NTA/9). Recruitment and data collection did not commence until January 2022 due to the COVID-19 pandemic. Data collection and analyses are expected to conclude in January 2024 and early 2025, respectively. Results are to be published in 2024 and 2025. CONCLUSIONS The results of this study will help us understand how the introduction of a specific prebiotic complementary food affects the microbiota and relative abundances of the microbial species, the modulation of immune development, and infant health. It will contribute to the expanding body of research that aims to deepen our understanding of the connections between nutrition, gut microbiota, and early-life postnatal health. TRIAL REGISTRATION Australian New Zealand Clinical Trials Registry ACTRN12620000026921; https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=378654. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/56772.
Collapse
Affiliation(s)
- Clare R Wall
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- Department of Nutrition, University of Otago, Dunedin, New Zealand
| | - Jane A Mullaney
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- AgResearch, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Warren Charles McNabb
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Olivier Gasser
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Karl Fraser
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- AgResearch, Palmerston North, New Zealand
| | - Eric Altermann
- BlueBarn Life Sciences Ltd., Palmerston North, New Zealand
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | | | - Janine Cooney
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- The New Zealand Institute for Plant and Food Research Ltd, Hamilton, New Zealand
| | - Robyn Lawrence
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Yannan Jiang
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Barbara C Galland
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Xiaoxi Fu
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Jacqueline N Tonkie
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand
| | - Nisha Mahawar
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
| | - Amy Luisa Lovell
- Department of Nutrition and Dietetics, University of Auckland, Auckland, New Zealand
- High-Value Nutrition, National Science Challenge, Auckland, New Zealand
| |
Collapse
|
3
|
Cruchet Muñoz S, Verbeke Palma S, Lera Marqués L, Espinosa Pizarro MN, Malig Mechasqui J, Sorensen K. Effects of Bifidobacterium longum 35624 in Children and Adolescents with Irritable Bowel Syndrome. Nutrients 2024; 16:1967. [PMID: 38931319 PMCID: PMC11206369 DOI: 10.3390/nu16121967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Irritable bowel syndrome (IBS) and vitamin D deficiency are common among children in Latin America. Previous studies show that Bifidobacterium longum35624TM improves IBS symptoms in adults. This real-world, single-arm, open-label study conducted in Chile investigated the effects of B. longum 35624 (1 × 109 colony-forming units, 12 weeks) on gastrointestinal symptoms (adapted IBS severity scoring system [IBS-SSS]; adapted Questionnaire on Pediatric Gastrointestinal Symptoms [QPGS], and Bristol Stool Form Scale) in 64 children and adolescents (8-18 years) and explored the relationship with baseline vitamin D status. Improvements in all IBS-SSS domains and composite score were observed at week 6 and 12 (p < 0.0007 versus baseline), with 98.3% of participants experiencing numerical improvements in ≥3 domains. Clinically meaningful improvement was seen in 96.6% of participants. The distribution of IBS-SSS severity categories shifted from moderate/severe at baseline to mild/remission (p < 0.0001). Improvements were not maintained during the two-week washout. Low baseline serum vitamin D levels did not correlate to IBS severity or probiotic response. QPGS significantly decreased from baseline to week 6 (p = 0.0005) and 12 (p = 0.02). B. longum 35624 may improve IBS symptoms in children and adolescents, even those with vitamin D deficiency. A confirmatory randomized controlled trial and further exploration of probiotic response and vitamin D status are needed.
Collapse
Affiliation(s)
- Sylvia Cruchet Muñoz
- Instituto de Nutrición y Tecnología de los Alimentos “Dr. Fernando Monckeberg”, Área Nutrición Humana, Universidad de Chile, El Líbano 5524, Macul, Santiago 7830490, Chile;
| | - Sandra Verbeke Palma
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Campus Santiago, Chile. Av. Ejército 146, Santiago 8370003, Chile
| | - Lydia Lera Marqués
- Faculty of Graduate Business and Education Programs, Keiser University eCampus, 1900 West Commercial Boulevard. Ste 100, Ft. Lauderdale, FL 33309, USA;
| | - María Nelly Espinosa Pizarro
- Facultad de Medicina, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago 7620001, Chile;
- Hospital Militar de Santiago, Av. Fernando Castillo Velasco 9100, La Reina, Santiago 7880047, Chile
| | | | - Katy Sorensen
- Medical Affairs, Novozymes A/S, Krogshøjvej 36, 2880 Bagsvaerd, Denmark;
| |
Collapse
|
4
|
Shaposhnikov LA, Tishkov VI, Pometun AA. Lactobacilli and Klebsiella: Two Opposites in the Fight for Human Health. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S71-S89. [PMID: 38621745 DOI: 10.1134/s0006297924140050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 04/17/2024]
Abstract
The problem of antibiotic resistance is currently very acute. Numerous research and development of new antibacterial drugs are being carried out that could help cope with various infectious agents. One of the promising directions for the search for new antibacterial drugs is the search among the probiotic strains present in the human gastrointestinal tract. This review is devoted to characteristics of one of these probiotic strains that have been studied to date: Limosilactobacillus reuteri. The review discusses its properties, synthesis of various compounds, as well as role of this strain in modulating various systems of the human body. The review also examines key characteristics of one of the most harmful among the currently known pathogenic organisms, Klebsiella, which is significantly resistant to antibiotics existing in medical practice, and also poses a great threat of nosocomial infections. Discussion of characteristics of the two strains, which have opposite effects on human health, may help in creation of new effective antibacterial drugs without significant side effects.
Collapse
Affiliation(s)
- Leonid A Shaposhnikov
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, 119071, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Vladimir I Tishkov
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, 119071, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Anastasia A Pometun
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, 119071, Russia.
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| |
Collapse
|
5
|
Dargenio VN, Cristofori F, Dargenio C, Giordano P, Indrio F, Celano G, Francavilla R. Use of Limosilactobacillus reuteri DSM 17938 in paediatric gastrointestinal disorders: an updated review. Benef Microbes 2022; 13:221-242. [PMID: 35212258 DOI: 10.3920/bm2021.0151] [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: 11/19/2022]
Abstract
Strains of lactobacilli are the most widely used probiotics and can be found in a large variety of food products and food supplements throughout the world. In this study, the evidence on Limosilactobacillus reuteri DSM 17938 (LR DSM 17938) has been reviewed. This species secretes reuterin and other substances singularly or in microvesicles, inhibiting pathogen growth and interacting with the intestinal microbiota and mucosa, restoring homeostasis. The use of LR DSM 17938 has been exploited in several pathological conditions. Preclinical research has shown that this probiotic can ameliorate dysbiosis and, by interacting with intestinal mucosal cells, can raise the pain threshold and promote gastrointestinal motility. These aspects are amongst the significant components in functional gastrointestinal disorders, such as colic and regurgitation in infants, functional abdominal pain and functional constipation in children and adolescents. This strain can decrease the duration of acute diarrhoea and hospitalization for acute gastroenteritis but does not seem to prevent nosocomial diarrhoea and antibiotic-associated diarrhoea. Because of its ability to survive in the gastric environment, it has been tested in Helicobacter pylori infection, showing a significant decrease of antibiotic-associated side effects and a tendency to increase the eradication rate. Finally, all these studies have shown the excellent safety of LR DSM 17938 even at higher dosages. In conclusion data from various clinical trials here reviewed can guide the clinician to find the correct dose, frequency of administration, and therapy duration.
Collapse
Affiliation(s)
- V N Dargenio
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - F Cristofori
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - C Dargenio
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - P Giordano
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - F Indrio
- Department of Paediatrics, University of Foggia, Via Pinto 1, 71100 Foggia, Italy
| | - G Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Via Amendola 265/a, 70126 Bari, Italy
| | - R Francavilla
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| |
Collapse
|
6
|
Imdad A, Rehman F, Davis E, Ranjit D, Surin GSS, Attia SL, Lawler S, Smith AA, Bhutta ZA. Effects of neonatal nutrition interventions on neonatal mortality and child health and development outcomes: A systematic review. CAMPBELL SYSTEMATIC REVIEWS 2021; 17:e1141. [PMID: 37133295 PMCID: PMC8356300 DOI: 10.1002/cl2.1141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background The last two decades have seen a significant decrease in mortality for children <5 years of age in low and middle-income countries (LMICs); however, neonatal (age, 0-28 days) mortality has not decreased at the same rate. We assessed three neonatal nutritional interventions that have the potential of reducing morbidity and mortality during infancy in LMICs. Objectives To determine the efficacy and effectiveness of synthetic vitamin A, dextrose oral gel, and probiotic supplementation during the neonatal period. Search Methods We conducted electronic searches for relevant studies on the following databases: PubMed, CINAHL, LILACS, SCOPUS, and CENTRAL, Cochrane Central Register for Controlled Trials, up to November 27, 2019. Selection Criteria We aimed to include randomized and quasi-experimental studies. The target population was neonates in LMICs. The interventions included synthetic vitamin A supplementation, oral dextrose gel supplementation, and probiotic supplementation during the neonatal period. We included studies from the community and hospital settings irrespective of the gestational age or birth weight of the neonate. Data Collection and Analysis Two authors screened the titles and extracted the data from selected studies. The risk of bias (ROB) in the included studies was assessed according to the Cochrane Handbook of Systematic Reviews. The primary outcome was all-cause mortality. The secondary outcomes were neonatal sepsis, necrotizing enterocolitis (NEC), prevention and treatment of neonatal hypoglycaemia, adverse events, and neurodevelopmental outcomes. Data were meta-analyzed by random effect models to obtain relative risk (RR) and 95% confidence interval (CI) for dichotomous outcomes and mean difference with 95% CI for continuous outcomes. The overall rating of evidence was determined by the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Main Results Sixteen randomized studies (total participants 169,366) assessed the effect of vitamin A supplementation during the neonatal period. All studies were conducted in low- and middle-income (LMIC) countries. Thirteen studies were conducted in the community setting and three studies were conducted in the hospital setting, specifically in neonatal intensive care units. Studies were conducted in 10 different countries including India (four studies), Guinea-Bissau (three studies), Bangladesh (two studies), and one study each in China, Ghana, Indonesia, Nepal, Pakistan, Tanzania, and Zimbabwe. The overall ROB was low in most of the included studies for neonatal vitamin A supplementation. The pooled results from the community based randomized studies showed that there was no significant difference in all-cause mortality in the vitamin A (intervention) group compared to controls at 1 month (RR, 0.99; 95% CI, 0.90-1.08; six studies with 126,548 participants, statistical heterogeneity I 2 0%, funnel plot symmetrical, grade rating high), 6 months (RR, 0.98; 95% CI, 0.89-1.07; 12 studies with 154,940 participants, statistical heterogeneity I 2 43%, funnel plot symmetrical, GRADE quality high) and 12 months of age (RR, 1.04; 95% CI, 0.94-1.14; eight studies with 118,376 participants, statistical heterogeneity I 2 46%, funnel plot symmetrical, GRADE quality high). Neonatal vitamin A supplementation increased the incidence of bulging fontanelle by 53% compared to control (RR, 1.53; 95% CI, 1.12-2.09; six studies with 100,256 participants, statistical heterogeneity I 2 65%, funnel plot symmetrical, GRADE quality high). We did not identify any experimental study that addressed the use of dextrose gel for the prevention and/or treatment of neonatal hypoglycaemia in LMIC. Thirty-three studies assessed the effect of probiotic supplementation during the neonatal period (total participants 11,595; probiotics: 5854 and controls: 5741). All of the included studies were conducted in LMIC and were randomized. Most of the studies were done in the hospital setting and included participants who were preterm (born < 37 weeks gestation) and/or low birth weight (<2500 g birth weight). Studies were conducted in 13 different countries with 10 studies conducted in India, six studies in Turkey, three studies each in China and Iran, two each in Mexico and South Africa, and one each in Bangladesh, Brazil, Colombia, Indonesia, Nepal, Pakistan, and Thailand. Three studies were at high ROB due to lack of appropriate randomization sequence or allocation concealment. Combined data from 25 studies showed that probiotic supplementation reduced all-cause mortality by 20% compared to controls (RR, 0.80; 95% CI, 0.66-0.96; total number of participants 10,998, number needed to treat 100, statistical heterogeneity I 2 0%, funnel plot symmetrical, GRADE quality high). Twenty-nine studies reported the effect of probiotics on the incidence of NEC, and the combined results showed a relative reduction of 54% in the intervention group compared to controls (RR, 0.46; 95% CI, 0.35-0.59; total number of participants 5574, number needed to treat 17, statistical heterogeneity I 2 24%, funnel plot symmetrical, GRADE quality high). Twenty-one studies assessed the effect of probiotic supplementation during the neonatal period on neonatal sepsis, and the combined results showed a relative reduction of 22% in the intervention group compared to controls (RR, 0.78; 95% CI, 0.70-0.86; total number of participants 9105, number needed to treat 14, statistical heterogeneity I 2 23%, funnel plot symmetrical, GRADE quality high). Authors' Conclusions Vitamin A supplementation during the neonatal period does not reduce all-cause neonatal or infant mortality in LMICs in the community setting. However, neonatal vitamin A supplementation increases the risk of Bulging Fontanelle. No experimental or quasi-experimental studies were available from LMICs to assess the effect of dextrose gel supplementation for the prevention or treatment of neonatal hypoglycaemia. Probiotic supplementation during the neonatal period seems to reduce all-cause mortality, NEC, and sepsis in babies born with low birth weight and/or preterm in the hospital setting. There was clinical heterogeneity in the use of probiotics, and we could not recommend any single strain of probiotics for wider use based on these results. There was a lack of studies on probiotic supplementation in the community setting. More research is needed to assess the effect of probiotics administered to neonates in-home/community setting in LMICs.
Collapse
Affiliation(s)
- Aamer Imdad
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and NutritionSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Faseeha Rehman
- Department of MedicineRaritan Bay Medical CenterPerth AmboyNew YorkUSA
| | - Evans Davis
- Roswell Park Comprehensive Cancer Center, Department of Cancer Prevention and ControlUniversity of BuffaloBuffaloNew YorkUSA
| | - Deepika Ranjit
- College of MedicineSUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | | | - Suzanna L. Attia
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and NutritionUniversity of KentuckyLexingtonKentuckyUSA
| | - Sarah Lawler
- Health Science LibrarySUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Abigail A. Smith
- Health Science LibraraySUNY Upstate Medical UniversitySyracuseNew YorkUSA
| | - Zulfiqar A. Bhutta
- Centre for Global Child HealthThe Hospital for Sick ChildrenTorontoOntarioCanada
| |
Collapse
|
7
|
Engevik MA, Ruan W, Esparza M, Fultz R, Shi Z, Engevik KA, Engevik AC, Ihekweazu FD, Visuthranukul C, Venable S, Schady DA, Versalovic J. Immunomodulation of dendritic cells by Lactobacillus reuteri surface components and metabolites. Physiol Rep 2021; 9:e14719. [PMID: 33463911 PMCID: PMC7814497 DOI: 10.14814/phy2.14719] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Lactic acid bacteria are commensal members of the gut microbiota and are postulated to promote host health. Secreted factors and cell surface components from Lactobacillus species have been shown to modulate the host immune system. However, the precise role of L. reuteri secreted factors and surface proteins in influencing dendritic cells (DCs) remains uncharacterized. HYPOTHESIS We hypothesize that L. reuteri secreted factors will promote DC maturation, skewing cells toward an anti-inflammatory phenotype. In acute colitis, we speculate that L. reuteri promotes IL-10 and dampens pro-inflammatory cytokine production, thereby improving colitis. METHODS & RESULTS Mouse bone marrow-derived DCs were differentiated into immature dendritic cells (iDCs) via IL-4 and GM-CSF stimulation. iDCs exposed to L. reuteri secreted factors or UV-irradiated bacteria exhibited greater expression of DC maturation markers CD83 and CD86 by flow cytometry. Additionally, L. reuteri stimulated DCs exhibited phenotypic maturation as denoted by cytokine production, including anti-inflammatory IL-10. Using mouse colonic organoids, we found that the microinjection of L. reuteri secreted metabolites and UV-irradiated bacteria was able to promote IL-10 production by DCs, indicating potential epithelial-immune cross-talk. In a TNBS-model of acute colitis, L. reuteri administration significantly improved histological scoring, colonic cytokine mRNA, serum cytokines, and bolstered IL-10 production. CONCLUSIONS Overall these data demonstrate that both L. reuteri secreted factors and its bacterial components are able to promote DC maturation. This work points to the specific role of L. reuteri in modulating intestinal DCs. NEW & NOTEWORTHY Lactobacillus reuteri colonizes the mammalian gastrointestinal tract and exerts beneficial effects on host health. However, the mechanisms behind these effects have not been fully explored. In this article, we identified that L. reuteri ATTC PTA 6475 metabolites and surface components promote dendritic cell maturation and IL-10 production. In acute colitis, we also demonstrate that L. reuteri can promote IL-10 and suppress inflammation. These findings may represent a crucial mechanism for maintaining intestinal immune homeostasis.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - Magdalena Esparza
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Robert Fultz
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Kristen A Engevik
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy C Engevik
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Faith D Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - Chonnikant Visuthranukul
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Pediatric Nutrition Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Susan Venable
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Deborah A Schady
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
8
|
Tan SK, Granados AC, Bouquet J, Hoy-Schulz YE, Green L, Federman S, Stryke D, Haggerty TD, Ley C, Yeh MT, Jannat K, Maldonado YA, Andino R, Parsonnet J, Chiu CY. Metagenomic sequencing of stool samples in Bangladeshi infants: virome association with poliovirus shedding after oral poliovirus vaccination. Sci Rep 2020; 10:15392. [PMID: 32958861 PMCID: PMC7506025 DOI: 10.1038/s41598-020-71791-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
The potential role of enteric viral infections and the developing infant virome in affecting immune responses to the oral poliovirus vaccine (OPV) is unknown. Here we performed viral metagenomic sequencing on 3 serially collected stool samples from 30 Bangladeshi infants following OPV vaccination and compared findings to stool samples from 16 age-matched infants in the United States (US). In 14 Bangladeshi infants, available post-vaccination serum samples were tested for polio-neutralizing antibodies. The abundance (p = 0.006) and richness (p = 0.013) of the eukaryotic virome increased with age and were higher than seen in age-matched US infants (p < 0.001). In contrast, phage diversity metrics remained stable and were similar to those in US infants. Non-poliovirus eukaryotic virus abundance (3.68 log10 vs. 2.25 log10, p = 0.002), particularly from potential viral pathogens (2.78log10 vs. 0.83log10, p = 0.002), and richness (p = 0.016) were inversely associated with poliovirus shedding. Following vaccination, 28.6% of 14 infants tested developed neutralizing antibodies to all three Sabin types and also exhibited higher rates of poliovirus shedding (p = 0.020). No vaccine-derived poliovirus variants were detected. These results reveal an inverse association between eukaryotic virome abundance and poliovirus shedding. Overall gut virome ecology and concurrent viral infections may impact oral vaccine responsiveness in Bangladeshi infants.
Collapse
Affiliation(s)
- Susanna K Tan
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrea C Granados
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Jerome Bouquet
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Yana Emmy Hoy-Schulz
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauri Green
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Scot Federman
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Doug Stryke
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Thomas D Haggerty
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Catherine Ley
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ming-Te Yeh
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Kaniz Jannat
- Environmental Intervention Unit, Infectious Disease Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Yvonne A Maldonado
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Julie Parsonnet
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA.
- Division of Infectious Diseases, Department of Medicine, University of California, 185 Berry Street, Box #0134, San Francisco, CA, 94107, USA.
| |
Collapse
|
9
|
Slums, Space, and State of Health-A Link between Settlement Morphology and Health Data. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17062022. [PMID: 32204347 PMCID: PMC7143924 DOI: 10.3390/ijerph17062022] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/31/2022]
Abstract
Approximately 1 billion slum dwellers worldwide are exposed to increased health risks due to their spatial environment. Recent studies have therefore called for the spatial environment to be introduced as a separate dimension in medical studies. Hence, this study investigates how and on which spatial scale relationships between the settlement morphology and the health status of the inhabitants can be identified. To this end, we summarize the current literature on the identification of slums from a geographical perspective and review the current literature on slums and health of the last five years (376 studies) focusing on the considered scales in the studies. We show that the majority of medical studies are restricted to certain geographical regions. It is desirable that the number of studies be adapted to the number of the respective population. On the basis of these studies, we develop a framework to investigate the relationship between space and health. Finally, we apply our methodology to investigate the relationship between the prevalence of slums and different health metrics using data of the global burden of diseases for different prefectures in Brazil on a subnational level.
Collapse
|
10
|
López L, Calderón D, Cardenas P, Prado MB, Valle C, Trueba G. Evolutionary changes of an intestinal Lactobacillus reuteri during probiotic manufacture. Microbiologyopen 2019; 9:e972. [PMID: 31746150 PMCID: PMC7002098 DOI: 10.1002/mbo3.972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 01/26/2023] Open
Abstract
Probiotic bacteria are frequently used to treat intestinal diseases or to improve health; however, little is known about the evolutionary changes of these bacteria during probiotic manufacture and the bacterial ability to colonize the intestine. It has been observed that when bacteria adapt to a new environment, they lose some traits required to thrive in the original niche. In this study, a strain of Lactobacillus reuteri was isolated from mouse duodenum and subjected to 150 serial passes in milk to simulate the industrial propagation of probiotic bacteria. The strains adapted to milk outperformed their ancestor when grown in milk; we also showed evidence of reduced intestinal colonization of milk‐adapted strains. Whole‐genome sequencing showed that bacterial adaptation to milk selects mutants with altered metabolic functions.
Collapse
Affiliation(s)
- Lázaro López
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Diana Calderón
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Paúl Cardenas
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - María B Prado
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Carlos Valle
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Gabriel Trueba
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| |
Collapse
|
11
|
Alexander LM, Oh JH, Stapleton DS, Schueler KL, Keller MP, Attie AD, van Pijkeren JP. Exploiting Prophage-Mediated Lysis for Biotherapeutic Release by Lactobacillus reuteri. Appl Environ Microbiol 2019; 85:e02335-18. [PMID: 30683744 PMCID: PMC6498169 DOI: 10.1128/aem.02335-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri has the potential to be developed as a microbial therapeutic delivery platform because of an established safety profile, health-promoting properties, and available genome editing tools. Here, we show that L. reuteri VPL1014 exhibits a low mutation rate compared to other Gram-positive bacteria, which we expect will contribute to the stability of genetically modified strains. VPL1014 encodes two biologically active prophages, which are induced during gastrointestinal transit. We hypothesized that intracellularly accumulated recombinant protein can be released following bacteriophage-mediated lysis. To test this, we engineered VPL1014 to accumulate leptin, our model protein, inside the cell. In vitro prophage induction of recombinant VPL1014 released leptin into the extracellular milieu, which corresponded to bacteriophage production. We also employed a plasmid system that does not require antibiotic in the growth medium for plasmid maintenance. Collectively, these data provide new avenues to exploit native prophages to deliver therapeutic molecules.IMPORTANCE Lactic acid bacteria (LAB) have been explored as potential biotherapeutic vehicles for the past 20 years. To secrete a therapeutic in the extracellular milieu, one typically relies on the bacterial secretion pathway, i.e., the Sec pathway. Overexpression of a secreted protein can overload the secretory pathway and impact the organism's fitness, and optimization of the signal peptide is also required to maximize the efficiency of the release of mature protein. Here, we describe a previously unexplored approach to release therapeutics from the probiotic Lactobacillus reuteri We demonstrate that an intracellularly accumulated recombinant protein is released following prophage activation. Since we recently demonstrated that prophages are activated during gastrointestinal transit, we propose that this method will provide a straightforward and efficient approach to deliver therapeutics in vivo.
Collapse
Affiliation(s)
- Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Donald S Stapleton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
12
|
Ong TG, Gordon M, Banks SSC, Thomas MR, Akobeng AK, Cochrane Developmental, Psychosocial and Learning Problems Group. Probiotics to prevent infantile colic. Cochrane Database Syst Rev 2019; 3:CD012473. [PMID: 30865287 PMCID: PMC6415699 DOI: 10.1002/14651858.cd012473.pub2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Infantile colic is typically defined as full-force crying for at least three hours per day, on at least three days per week, for at least three weeks. Infantile colic affects a large number of infants and their families worldwide. Its symptoms are broad and general, and while not indicative of disease, may represent a serious underlying condition in a small percentage of infants who may need a medical assessment. Probiotics are live microorganisms that alter the microflora of the host and provide beneficial health effects. The most common probiotics used are of Lactobacillus, Bifidobacterium and Streptococcus. There is growing evidence to suggest that intestinal flora in colicky infants differ from those in healthy infants, and it is suggested that probiotics can redress this balance and provide a healthier intestinal microbiota landscape. The low cost and easy availability of probiotics makes them a potential prophylactic solution to reduce the incidence and prevalence of infantile colic. OBJECTIVES To evaluate the efficacy and safety of prophylactic probiotics in preventing or reducing severity of infantile colic. SEARCH METHODS In January 2018 we searched CENTRAL, MEDLINE, Embase, PsycINFO, CINAHL, 10 other databases and two trials registers. In addition, we handsearched the abstracts of relevant meetings, searched reference lists, ran citation searches of included studies, and contacted authors and experts in the field, including the manufacturers of probiotics, to identify unpublished trials. SELECTION CRITERIA Randomised control trials (RCTs) of newborn infants less than one month of age without the diagnosis of infantile colic at recruitment. We included any probiotic, alone or in combination with a prebiotic (also known as synbiotics), versus no intervention, another intervention(s) or placebo, where the focus of the study was the effect of the intervention on infantile colic. DATA COLLECTION AND ANALYSIS We used standard methodological procedures of Cochrane. MAIN RESULTS Our search yielded 3284 records, and of these, we selected 21 reports for full-text review. Six studies with 1886 participants met our inclusion criteria, comparing probiotics with placebo. Two studies examined Lactobacillus reuteri DSM, two examined multi-strain probiotics, one examined Lactobacillus rhamnosus, and one examined Lactobacillus paracasei and Bifidobacterium animalis. Two studies began probiotics during pregnancy and continued administering them to the baby after birth.We considered the risk of bias for randomisation as low for all six trials; for allocation concealment as low in two studies and unclear in four others. All studies were blinded, and at low risk of attrition and reporting bias.A random-effects meta-analysis of three studies (1148 participants) found no difference between the groups in relation to occurrence of new cases of colic: risk ratio (RR) 0.46, 95% confidence interval (CI) 0.18 to 1.19; low-certainty evidence; I2 = 72%.A random-effects meta-analysis of all six studies (1851 participants) found no difference between the groups in relation to serious adverse effects (RR 1.02, 95% CI 0.14 to 7.21; low-certainty evidence; I2 not calculable (only four serious events for one comparison, two in each group: meconium plug obstruction, patent ductus arteriosus and neonatal hepatitis).A random-effects meta-analysis of three studies (707 participants) found a mean difference (MD) of -32.57 minutes per day (95% CI -55.60 to -9.54; low-certainty evidence; I2 = 93%) in crying time at study end in favour of probiotics.A subgroup analysis of the most studied agent, Lactobacillus reuteri, showed a reduction of 44.26 minutes in daily crying with a random-effects model (95% CI -66.6 to -21.9; I2 = 92%), in favour of probiotics. AUTHORS' CONCLUSIONS There is no clear evidence that probiotics are more effective than placebo at preventing infantile colic; however, daily crying time appeared to reduce with probiotic use compared to placebo. There were no clear differences in adverse effects.We are limited in our ability to draw conclusions by the certainty of the evidence, which we assessed as being low across all three outcomes, meaning that we are not confident that these results would not change with the addition of further research.
Collapse
Affiliation(s)
- Teck Guan Ong
- Blackpool Victoria HospitalChild Health DepartmentWhinney Heys RoadBlackpoolLancashireUKFY3 8NR
| | - Morris Gordon
- University of Central LancashireSchool of MedicinePrestonLancashireUK
- Blackpool Victoria HospitalFamilies DivisionBlackpoolUK
| | - Shel SC Banks
- Blackpool Teaching Hospitals NHS Foundation TrustDepartment of Child HealthWhinney Heys RoadBlackpoolUKFY3 8NR
| | - Megan R Thomas
- Blackpool Teaching Hospitals NHS Foundation TrustDepartment of Child HealthWhinney Heys RoadBlackpoolUKFY3 8NR
- Lancaster UniversityFaculty of Health and MedicineFurness CollegeLancasterUKLA1 4YG
| | - Anthony K Akobeng
- Sidra MedicinePO Box 26999DohaQatar
- Cornell UniversityWeill Cornell MedicineDohaQatar
| | | |
Collapse
|
13
|
Roberts TJ, Hoy-Schulz YE, Jannat K, Parsonnet J. Evidence of inflated exclusive breastfeeding estimates from a clinical trial in Bangladesh. Int Breastfeed J 2018; 13:39. [PMID: 30159001 PMCID: PMC6106757 DOI: 10.1186/s13006-018-0179-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/30/2018] [Indexed: 11/15/2022] Open
Abstract
Suboptimal breastfeeding is a major cause of infant morbidity and mortality across the world. Inconsistent data has hampered quantification of this practice, however, limiting breastfeeding promotion efforts. As part of a clinical trial in Dhaka, Bangladesh, data was collected on breastfeeding patterns among 125 infants. Infants were ages 4 to 12 weeks (mean = 8.05, SD = 2.13) at the time of enrollment, and breastfeeding data were collected at 24 study visits during a twelve-week period. Breastfeeding status was assessed using the WHO-recommended “current status” (24-h recall) method. These data were used to calculate two measures: a longitudinal estimate of exclusive breastfeeding since birth and a simulated cross-sectional prevalence to approximate common data collection methods. Infants were then ranked based on their breastfeeding status at all study visits and grouped into quartiles and compared using hospitalization data recorded for all infants as part of the original study. These data showed large differences in estimates of exclusive breastfeeding behaviors when assessed longitudinally (8.8% exclusive breastfeeding) vs. calculating a cross-sectional prevalence (56.2% exclusive breastfeeding). Additionally, when infants were grouped by quartile of breastfeeding behavior and matched with hospitalization records, it was found that infants in the lowest quartile of breastfeeding behaviors were significantly more likely to be hospitalized than infants in the highest quartile. These results provide further evidence that current breastfeeding epidemiology studies may overestimate rates of exclusive breastfeeding. They also provide further evidence to support the significant infant health benefits from breastfeeding promotion. Trial registration: ClinicalTrials.gov NCT01899378. Registered July 10, 2013.
Collapse
Affiliation(s)
- Thomas J Roberts
- 1Department of Medicine, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114 USA
| | - Yana E Hoy-Schulz
- 2Department of Medicine, Stanford University School of Medicine, Stanford, USA
| | - Kaniz Jannat
- 3Environmental Intervention Unit, Infectious Disease Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Julie Parsonnet
- 2Department of Medicine, Stanford University School of Medicine, Stanford, USA.,4Department of Health Research and Policy, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
14
|
Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in Human Health and Diseases. Front Microbiol 2018; 9:757. [PMID: 29725324 PMCID: PMC5917019 DOI: 10.3389/fmicb.2018.00757] [Citation(s) in RCA: 463] [Impact Index Per Article: 66.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri (L. reuteri) is a well-studied probiotic bacterium that can colonize a large number of mammals. In humans, L. reuteri is found in different body sites, including the gastrointestinal tract, urinary tract, skin, and breast milk. The abundance of L. reuteri varies among different individuals. Several beneficial effects of L. reuteri have been noted. First, L. reuteri can produce antimicrobial molecules, such as organic acids, ethanol, and reuterin. Due to its antimicrobial activity, L. reuteri is able to inhibit the colonization of pathogenic microbes and remodel the commensal microbiota composition in the host. Second, L. reuteri can benefit the host immune system. For instance, some L. reuteri strains can reduce the production of pro-inflammatory cytokines while promoting regulatory T cell development and function. Third, bearing the ability to strengthen the intestinal barrier, the colonization of L. reuteri may decrease the microbial translocation from the gut lumen to the tissues. Microbial translocation across the intestinal epithelium has been hypothesized as an initiator of inflammation. Therefore, inflammatory diseases, including those located in the gut as well as in remote tissues, may be ameliorated by increasing the colonization of L. reuteri. Notably, the decrease in the abundance of L. reuteri in humans in the past decades is correlated with an increase in the incidences of inflammatory diseases over the same period of time. Direct supplementation or prebiotic modulation of L. reuteri may be an attractive preventive and/or therapeutic avenue against inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
15
|
Xu X, Jia X, Mo L, Liu C, Zheng L, Yuan Q, Zhou X. Intestinal microbiota: a potential target for the treatment of postmenopausal osteoporosis. Bone Res 2017; 5:17046. [PMID: 28983411 PMCID: PMC5627629 DOI: 10.1038/boneres.2017.46] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Postmenopausal osteoporosis (PMO) is a prevalent metabolic bone disease characterized by bone loss and structural destruction, which increases the risk of fracture in postmenopausal women. Owing to the high morbidity and serious complications of PMO, many efforts have been devoted to its prophylaxis and treatment. The intestinal microbiota is the complex community of microorganisms colonizing the gastrointestinal tract. Probiotics, which are dietary or medical supplements consisting of beneficial intestinal bacteria, work in concert with endogenous intestinal microorganisms to maintain host health. Recent studies have revealed that bone loss in PMO is closely related to host immunity, which is influenced by the intestinal microbiota. The curative effects of probiotics on metabolic bone diseases have also been demonstrated. The effects of the intestinal microbiota on bone metabolism suggest a promising target for PMO management. This review seeks to summarize the critical effects of the intestinal microbiota and probiotics on PMO, with a focus on the molecular mechanisms underlying the pathogenic relationship between bacteria and host, and to define the possible treatment options.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyue Jia
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Longyi Mo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Dental Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Lyseng-Williamson KA. Bifidobacterium infantis 35624 as a probiotic dietary supplement: a profile of its use. DRUGS & THERAPY PERSPECTIVES 2017. [DOI: 10.1007/s40267-017-0423-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Smilowitz JT, Moya J, Breck MA, Cook C, Fineberg A, Angkustsiri K, Underwood MA. Safety and tolerability of Bifidobacterium longum subspecies infantis EVC001 supplementation in healthy term breastfed infants: a phase I clinical trial. BMC Pediatr 2017; 17:133. [PMID: 28558732 PMCID: PMC5450358 DOI: 10.1186/s12887-017-0886-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/17/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Historically, bifidobacteria were the dominant intestinal bacteria in breastfed infants. Still abundant in infants in developing nations, levels of intestinal bifidobacteria are low among infants in developed nations. Recent studies have described an intimate relationship between human milk and a specific subspecies of Bifidobacterium, B. longum subsp. infantis (B. infantis), yet supplementation of breastfed, healthy, term infants with this organism, has not been reported. The IMPRINT Study, a Phase I clinical trial, was initiated to determine the safety and tolerability of supplementing breastfed infants with B. infantis (EVC001). METHODS Eighty mother-infant dyads were enrolled in either lactation support plus B. infantis supplementation (BiLS) or lactation support alone (LS). Starting with Day 7 postnatal, BiLS infants were fed 1.8-2.8 × 1010 CFU B. infantis EVC001 daily in breast milk for 21 days. Mothers collected fecal samples, filled out health questionnaires, and kept daily logs about their infants' feeding and gastrointestinal symptoms from birth until Day 61 postnatal. Safety and tolerability were determined from maternal reports. RESULTS There were no differences in the mean gestational age at birth, weight 1 and 2 months postnatal, and breast milk intake between groups. The mean Log10 change in fecal Bifidobacterium from Day 6 to Day 28 was higher (p = 0.0002) for BiLS (6.6 ± 2.8 SD) than for LS infants (3.5 ± 3.5 SD). Daily stool number was higher (p < 0.005) for LS and lower (p < 0.05) for BiLS infants during supplementation than at Baseline. During supplementation, watery stools decreased and soft stools increased by 36% over baseline in BiLS infants (p < 0.05) with no significant changes in stool consistency for the LS infants. None of the safety and tolerability endpoints, including flatulence, bloody stool, body temperature, ratings of gastrointestinal symptoms, use of antibiotics or gas-relieving medications, infant colic, jaundice, number of illnesses, sick doctor visits, or diagnoses of eczema were different for the groups at any point. CONCLUSIONS The B. infantis EVC001 supplement was safely consumed and well-tolerated. Stools were fewer and better formed in infants in the BiLS group compared with LS group. Adverse events were those expected in healthy infants and not different between groups. TRIAL REGISTRATION ClinicalTrials.gov NCT02457338 . Registered May 27, 2015.
Collapse
Affiliation(s)
- Jennifer T. Smilowitz
- Department of Food Science and Technology, University of California, Davis, CA USA
- Foods for Health Institute, University of California, One Shields Ave, Davis, CA 95616 USA
| | - Jackelyn Moya
- Department of Food Science and Technology, University of California, Davis, CA USA
| | - Melissa A. Breck
- Department of Food Science and Technology, University of California, Davis, CA USA
| | - Chelsea Cook
- Department of Food Science and Technology, University of California, Davis, CA USA
| | | | | | - Mark A. Underwood
- Department of Pediatrics, UC Davis Children’s Hospital, Sacramento, CA USA
| |
Collapse
|