1
|
Vievermanns K, Dierikx TH, Oldenburger NJ, Jamaludin FS, Niemarkt HJ, de Meij TGJ. Effect of probiotic supplementation on the gut microbiota in very preterm infants: a systematic review. Arch Dis Child Fetal Neonatal Ed 2024; 110:57-67. [PMID: 38925919 DOI: 10.1136/archdischild-2023-326691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/13/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE There is increasing evidence that probiotic supplementation in very preterm infants decreases the risk of necrotising enterocolitis (NEC), sepsis and mortality. The underlying mechanisms, including effects on the gut microbiota, are largely unknown. We aimed to systematically review the available literature on the effects of probiotic supplementation in very preterm infants on gut microbiota development. DESIGN A systematic review in Medline, Embase, Cochrane Library, CINAHL and Web of Science. SETTING Neonatal intensive care unit. PATIENTS Premature infants. INTERVENTION Probiotic supplementation. MAIN OUTCOME MEASURES Gut microbiota. RESULTS A total of 1046 articles were screened, of which 29 were included. There was a large heterogeneity in study design, dose and type of probiotic strains, timepoints of sample collection and analysing techniques. Bifidobacteria and lactobacilli were the most used probiotic strains. The effects of probiotics on alpha diversity were conflicting; however, beta diversity was significantly different between probiotic-supplemented infants and controls in the vast majority of studies. In most studies, probiotic supplementation led to increased relative abundance of the supplemented strains and decreased abundance of genera such as Clostridium, Streptococcus, Klebsiella and Escherichia. CONCLUSIONS Probiotic supplementation to preterm infants seems to increase the relative abundance of the supplemented strains with a concurrent decrease of potentially pathogenic species. These probiotic-induced microbial alterations may contribute to the decreased risk of health complications such as NEC. Future trials, including omics technologies to analyse both microbiota composition and function linked to health outcomes, are warranted to identify the optimal mixture and dosing of probiotic strains. PROSPERO REGISTRATION NUMBER CRD42023385204.
Collapse
Affiliation(s)
- Kayleigh Vievermanns
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Thomas H Dierikx
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Microbiology, Maastricht UMC+, Maastricht, The Netherlands
| | | | - Faridi S Jamaludin
- Medical Library AMC, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Hendrik J Niemarkt
- Neonatology, Maxima Medisch Centrum locatie Veldhoven, Veldhoven, The Netherlands
- Electrical Engineering, TU Eindhoven, Eindhoven, The Netherlands
| | - Tim G J de Meij
- Pediatric Gastroenterology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Pediatric Gastroenterology, Emma children's hospital amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Wang L, Zhang Z, Zhu X, Zhao Y, Iqbal M, Lin Z, Nawaz S, Xu M, Hu M, Bhutto ZA, Li J. The Effect of Lactobacillus sakei on Growth Performance and Intestinal Health in Dogs: Gut Microbiota and Metabolism Study. Probiotics Antimicrob Proteins 2024; 16:2116-2131. [PMID: 37740881 DOI: 10.1007/s12602-023-10160-9] [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] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
The gut microbiota is the largest and most complex ecosystem consisting of trillions of microorganisms, which influenced by various external factors. As an important probiotic species, Lactobacillus helps to improve gut microbial diversity and composition, underlying potential efficacy in growth performance and disease prevention. However, limited studies have been investigated the relationship between Lactobacillus sakei and intestinal health in dogs. In this study, dogs in the two groups were fed a standard diet (group C, n = 8) and Lactobacillus sakei diet (group P, n = 8), respectively. The growth performance, serum biochemical indices, antioxidant capacity, gut microbiota, and metabolism of dogs in both groups were studied. Results from growth trials showed that L. sakei can significantly improve the growth performance of dogs, including increased weight gain (p < 0.05), serum biochemical indices, i.e., ALP, TP, and ALB (p < 0.05), and better antioxidant capacity, i.e., SOD and GSH-Px (p < 0.05). Significant changes in the gut microbial composition were detected in dogs fed Lactobacillus sakei, as evidenced by an increase in the level of Firmicutes, Spirochaetota, and Patescibacteria, all of them play an important role in maintaining intestinal health. Moreover, a decrease in the level of microorganisms that threaten health, such as Mucispirillum and Clostridium_sensu_stricto_13. The metabolic analysis showed that the Lactobacillus sakei enhanced metabolic pathways such as vitamin B6 metabolism, glutathione metabolism, retinol metabolism, and fatty acid degradation. Our findings suggested that Lactobacillus sakei supplementation had beneficial effects on the growth performance and health status of dogs by improving gut microbiota balance and promoting metabolism. There are an estimated 200 million dogs in China, and the population is continuing to grow at a rapid pace. It is essential to explore an effective way to promote health in dogs. Intestinal diseases, particularly colitis and diarrhea, are common clinical conditions in dogs and are associated with gut microbiota. Lactobacillus sakei, as an important species of probiotics, the relationship between L. sakei and intestinal health in dogs remains unclear. Our study suggests that L. sakei significantly promotes growth performance and health states involving weight gain, regulation of gut microbiota, and metabolism. Overall, our findings shed light on the potential role of L. sakei as an alternative in promoting health in dogs.
Collapse
Affiliation(s)
- Lei Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhenwen Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210000, China
| | - Xiaohui Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuanfeng Zhao
- Institute of Animal Husbandry and Veterinary Science, Guizhou Academy of Agricultural Sciences, Guiyang, 550005, Guizhou, China
| | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhengrong Lin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mengen Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Miao Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zohaib Ahmed Bhutto
- Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water and Marine Science, Uthal, Pakistan
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
3
|
Raeisi H, Leeflang J, Hasan S, Woods SL. Bioengineered Probiotics for Clostridioides difficile Infection: An Overview of the Challenges and Potential for This New Treatment Approach. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10398-x. [PMID: 39531149 DOI: 10.1007/s12602-024-10398-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The rapid increase in microbial antibiotic resistance in Clostridioides difficile (C. difficile) strains and the formation of hypervirulent strains have been associated with a global increase in the incidence of C. difficile infection (CDI) and subsequently, an increase in the rate of recurrence. These consequences have led to an urgent need to develop new and promising alternative strategies to control this pathogen. Engineered probiotics are exciting new bacterial strains produced by editing the genome of the original probiotics. Recently, engineered probiotics have been used to develop delivery vehicles for vaccines, diagnostics, and therapeutics. Recent studies have demonstrated engineered probiotics may potentially be an effective approach to control or treat CDI. This review provides a brief overview of the considerations for engineered probiotics for medicinal use, with a focus on recent preclinical research using engineered probiotics to prevent or treat CDI. We also address the challenges faced in the production of engineered strains and how they may be overcome in the application of these agents to meet patient needs in the future.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Julia Leeflang
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Sadia Hasan
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Susan L Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| |
Collapse
|
4
|
Qazi KR, Govindaraj D, Martí M, de Jong Y, Jensen GB, Abrahamsson T, Jenmalm MC, Sverremark-Ekström E. Impact of Extreme Prematurity, Chorioamnionitis, and Sepsis on Neonatal Monocyte Characteristics and Functions. J Innate Immun 2024; 16:470-488. [PMID: 39278208 PMCID: PMC11521501 DOI: 10.1159/000541468] [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: 04/19/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION The innate branch of the immune system is important in early life, in particular for infants born preterm. METHODS We performed a longitudinal analysis of the peripheral monocyte compartment in extremely preterm children from a randomized, placebo-controlled study of probiotic supplementation. PBMCs and fecal samples were collected at several timepoints during the first months of life. Monocyte characteristics were analyzed by flow cytometry, and LPS-stimulated PBMC culture supernatants were analyzed by Luminex or ELISA. Plasma cytokines and gut microbiota composition were analyzed by ELISA and 16S rRNA-sequencing, respectively. RESULTS The extremely preterm infants had persistent alterations in their monocyte characteristics that were further aggravated in chorioamnionitis cases. They showed a markedly reduced TLR4 expression and hampered LPS-stimulated cytokine responses 14 days after birth. Notably, at later timepoints, TLR4 expression and LPS responses no longer correlated. Sepsis during the first weeks of life strongly associated with increased pro-inflammatory, and reduced IL-10, responses also at postmenstrual week 36. Further, we report a correlation between gut microbiota features and monocyte phenotype and responses, but also that probiotic supplementation associated with distinct monocyte phenotypic characteristics, without significantly influencing their responsiveness. CONCLUSION Extremely preterm infants have monocyte characteristics and functional features that deviate from infants born full-term. Some of these differences persist until they reach an age corresponding to full-term, potentially making them more vulnerable to microbial exposures during the first months of life.
Collapse
Affiliation(s)
- Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Dhanapal Govindaraj
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Magalí Martí
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ymke de Jong
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Georg Bach Jensen
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Thomas Abrahamsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Crown Princess Victoria Children’s Hospital, Linköping, Sweden
| | - Maria C. Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
5
|
Wang L, Ren B, Wu S, Song H, Xiong L, Wang F, Shen X. Current research progress, opportunities, and challenges of Limosillactobacillus reuteri-based probiotic dietary strategies. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38920093 DOI: 10.1080/10408398.2024.2369946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Limosillactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., stands out as the most extensively researched probiotic. Its remarkable intestinal adhesion has led to widespread applications in both the food and medical sectors. Notably, recent research highlights the probiotic efficacy of L. reuteri sourced from breast milk, particularly in influencing social behavior and mitigating atopic dermatitis. In this review, our emphasis is on surveying recent literature regarding the promotion of host's health by L. reuteri. We aim to provide a concise summary of the latest regulatory effects and potential mechanisms attributed to L. reuteri in the realms of metabolism, brain- and immune-related functions. The mechanism through which L. reuteri promotes host health by modulating the intestinal microenvironment primarily involves promoting intestinal epithelial renewal, bolstering intestinal barrier function, regulating gut microbiota and its metabolites, and suppressing inflammation and immune responses. Additionally, this review delves into new technologies, identifies shortcomings, and addresses challenges in current L. reuteri research. Finally, the application prospects of L. reuteri are provided. Therefore, a better understanding of the role and mechanisms of L. reuteri will contribute significantly to the development of new probiotic functional foods and enable precise, targeted interventions for various diseases.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Shufeng Wu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
6
|
Chen W, Guo K, Huang X, Zhang X, Li X, Chen Z, Wang Y, Wang Z, Liu R, Qiu H, Wang M, Zeng S. The Association of Neonatal Gut Microbiota Community State Types with Birth Weight. CHILDREN (BASEL, SWITZERLAND) 2024; 11:770. [PMID: 39062221 PMCID: PMC11276374 DOI: 10.3390/children11070770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND while most gut microbiota research has focused on term infants, the health outcomes of preterm infants are equally important. Very-low-birth-weight (VLBW) or extremely-low-birth-weight (ELBW) preterm infants have a unique gut microbiota structure, and probiotics have been reported to somewhat accelerate the maturation of the gut microbiota and reduce intestinal inflammation in very-low preterm infants, thereby improving their long-term outcomes. The aim of this study was to investigate the structure of gut microbiota in ELBW neonates to facilitate the early identification of different types of low-birth-weight (LBW) preterm infants. METHODS a total of 98 fecal samples from 39 low-birth-weight preterm infants were included in this study. Three groups were categorized according to different birth weights: ELBW (n = 39), VLBW (n = 39), and LBW (n = 20). The gut microbiota structure of neonates was obtained by 16S rRNA gene sequencing, and microbiome analysis was conducted. The community state type (CST) of the microbiota was predicted, and correlation analysis was conducted with clinical indicators. Differences in the gut microbiota composition among ELBW, VLBW, and LBW were compared. The value of gut microbiota composition in the diagnosis of extremely low birth weight was assessed via a random forest-machine learning approach. RESULTS we briefly analyzed the structure of the gut microbiota of preterm infants with low birth weight and found that the ELBW, VLBW, and LBW groups exhibited gut microbiota with heterogeneous compositions. Low-birth-weight preterm infants showed five CSTs dominated by Enterococcus, Staphylococcus, Klebsiella, Streptococcus, Pseudescherichia, and Acinetobacter. The birth weight and clinical indicators related to prematurity were associated with the CST. We found the composition of the gut microbiota was specific to the different types of low-birth-weight premature infants, namely, ELBW, VLBW, and LBW. The ELBW group exhibited significantly more of the potentially harmful intestinal bacteria Acinetobacter relative to the VLBW and LBW groups, as well as a significantly lower abundance of the intestinal probiotic Bifidobacterium. Based on the gut microbiota's composition and its correlation with low weight, we constructed random forest model classifiers to distinguish ELBW and VLBW/LBW infants. The area under the curve of the classifiers constructed with Enterococcus, Klebsiella, and Acinetobacter was found to reach 0.836 by machine learning evaluation, suggesting that gut microbiota composition may be a potential biomarker for ELBW preterm infants. CONCLUSIONS the gut bacteria of preterm infants showed a CST with Enterococcus, Klebsiella, and Acinetobacter as the dominant genera. ELBW preterm infants exhibit an increase in the abundance of potentially harmful bacteria in the gut and a decrease in beneficial bacteria. These potentially harmful bacteria may be potential biomarkers for ELBW preterm infants.
Collapse
Affiliation(s)
- Wanling Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518116, China
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
| | - Kaiping Guo
- Division of Pediatrics, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xueli Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Xiaoxia Li
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Zimiao Chen
- Department of Burn Plastic Surgery, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Yanli Wang
- Department of Pediatrics, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Rongtian Liu
- Department of Pediatrics, Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
- Department of Neonatology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen 518172, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| |
Collapse
|
7
|
TAKESHITA K, TAKEI H, TANAKA S, HISHIKI H, IIJIMA Y, OGATA H, FUJISHIRO K, TOMINAGA T, KONNO Y, IWASE Y, ENDO M, ISHIWADA N, OSONE Y, TAKEMURA R, HAMADA H, SHIMOJO N. Effect of multi-strain bifidobacteria supplementation on intestinal microbiota development in low birth weight neonates: a randomized controlled trial. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:352-358. [PMID: 39364130 PMCID: PMC11444860 DOI: 10.12938/bmfh.2023-093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/21/2024] [Indexed: 10/05/2024]
Abstract
Single-strain Bifidobacterium species are commonly used as probiotics with low birth weight neonates. However, the effectiveness and safety of multi-strain Bifidobacterium supplementation are not well known. Thirty-six neonates weighing less than 2,000 g (558-1,943 g) at birth and admitted to a neonatal intensive care unit were randomly assigned to receive a single strain or triple strains of Bifidobacterium with lactulose enterally for 4 weeks from birth. The relative abundances of Staphylococcus and Bifidobacterium in the fecal microbiota at weeks 1, 2, and 4 were investigated. Based on the study results, no significant difference was detected between the two groups in the abundance of Staphylococcus; however, the triple-strain group had significantly high abundances of Bifidobacterium at weeks 2 and 4. The fecal microbiota in the triple-strain group had significantly lower alpha diversity (Bifidobacterium-enriching) after week 4 and was different from that in the single-strain group, which showed a higher abundance of Clostridium. No severe adverse events occurred in either group during the study period. Although no significant difference was detected between single- and multi-strain bifidobacteria supplementation in the colonization of Staphylococcus in the fecal microbiota of the neonates, multi-strain bifidobacteria supplementation contributed toward early enrichment of the microbiota with bifidobacteria and suppression of other pathogenic bacteria, such as Clostridium spp.
Collapse
Affiliation(s)
- Kenichi TAKESHITA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Department of Pediatrics, Chiba Rosai Hospital, 2-16
Tatsumidaihigashi, Ichihara-shi, Chiba 290-0003, Japan
| | - Haruka TAKEI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Saori TANAKA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Haruka HISHIKI
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Yuta IIJIMA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Hitoshi OGATA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Kensuke FUJISHIRO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Takahiro TOMINAGA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yuki KONNO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Yukiko IWASE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Mamiko ENDO
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Naruhiko ISHIWADA
- Department of Infectious Diseases, Medical Mycology Research
Center, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8673, Japan
| | - Yoshiteru OSONE
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
- Perinatal Medical Center, Chiba University Hospital, 1-8-1
Inohana, Chuo-ku, Chiba-shi, Chiba 260-8677, Japan
| | - Ryo TAKEMURA
- Clinical and Translational Research Center, Keio University
Hospital, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiromichi HAMADA
- Department of Pediatrics, Graduate School of Medicine, Chiba
University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| | - Naoki SHIMOJO
- Center for Preventive Medical Sciences, Chiba University,
1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan
| |
Collapse
|
8
|
Govindaraj D, Jensen GB, Rahman Qazi K, Sverremark‐Ekström E, Abrahamsson T, Jenmalm MC. Effects of extremely preterm birth on cytokine and chemokine responses induced by T-cell activation during infancy. Clin Transl Immunology 2024; 13:e1510. [PMID: 38737447 PMCID: PMC11087183 DOI: 10.1002/cti2.1510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
Objectives Extremely preterm (EPT; gestational week < 28 + 0, < 1000 g) neonates are vulnerable to infections and necrotising enterocolitis, important contributors to mortality and morbidity. However, knowledge regarding their immune maturation remains limited. We here investigated the longitudinal development of functional T-cell capacity in EPT infants. Methods Peripheral blood mononuclear cells were isolated at 14th and 28th day (D) and at gestational week 36 + 0 (Gw36) from EPT infants, participated in a randomised, double-blind, placebo-controlled study of Lactobacillus reuteri DSM 17938 probiotic supplementation. Blood collected from 25 full-term (FT) infants at D14 was used as control. The secretion of immune mediators was determined through comprehensive Luminex panels after stimulation with human T-cell activator CD3/CD28 beads. Results The levels of many mediators were low in EPT infants at D14, whereas the secretion of several chemokines was higher in EPT than in FT infants. Furthermore, Th2:Th1 cytokine ratios were higher in EPT than in FT infants. Progressively elevated secretion of, for example, IFN-γ, TNF and IL-17A in EPT infants was observed from D14 to D28 and then at Gw36. Elevated levels were observed for many proinflammatory mediators at D28. Probiotic supplementation or perinatal factors (e.g. clinical chorioamnionitis, preeclampsia and delivery mode) did not influence the cytokine and chemokine responses. Conclusions Immune mediators induced by T-cell activation in EPT infants were mainly reduced at D14 and Th2 skewed compared to those in FT infants, but mostly recovered at Gw36, indicating immune maturation. Increased proinflammatory responses at D28 may be related to the heightened risk of severe immune-associated complications seen in EPT infants.
Collapse
Affiliation(s)
- Dhanapal Govindaraj
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| | - Georg Bach Jensen
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Eva Sverremark‐Ekström
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Thomas Abrahamsson
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| |
Collapse
|
9
|
Wala SJ, Ragan MV, Pryor E, Canvasser J, Diefenbach KA, Besner GE. Contemporary use of prophylactic probiotics in NICUs in the United States: a survey update. J Perinatol 2024; 44:739-744. [PMID: 38553600 DOI: 10.1038/s41372-024-01952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 05/15/2024]
Abstract
OBJECTIVE In 2015, 14.0% of US NICUs administered probiotics to very low birth weight infants. Current probiotic use prior to and after the Fall of 2023 (when FDA warnings were issued) remains unknown. STUDY DESIGN A survey was distributed to the American Academy of Pediatrics Section on Neonatal and Perinatal Medicine (August-November/2022) and Neonatology Solutions' Level III/IV NICUs (January-April/2023). Probiotic administration practices were investigated. RESULTS In total, 289 unique NICUs and 406 providers responded to the survey. Of those, 29.1% of NICUs administered prophylactic probiotics to premature neonates, however, this decreased considerably after FDA warnings were issued. Additionally, 71.4% of providers stated willingness to administer probiotics to premature infants if there was an FDA-approved formulation. CONCLUSIONS Probiotic use in US NICUs increased between 2015 and the Fall of 2023 and then dropped dramatically following warning letters from the FDA. The introduction of an FDA-approved probiotic may further expand administration.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | - Karen A Diefenbach
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
10
|
Josephs-Spaulding J, Rajput A, Hefner Y, Szubin R, Balasubramanian A, Li G, Zielinski DC, Jahn L, Sommer M, Phaneuf P, Palsson BO. Reconstructing the transcriptional regulatory network of probiotic L. reuteri is enabled by transcriptomics and machine learning. mSystems 2024; 9:e0125723. [PMID: 38349131 PMCID: PMC10949432 DOI: 10.1128/msystems.01257-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/09/2024] [Indexed: 03/20/2024] Open
Abstract
Limosilactobacillus reuteri, a probiotic microbe instrumental to human health and sustainable food production, adapts to diverse environmental shifts via dynamic gene expression. We applied the independent component analysis (ICA) to 117 RNA-seq data sets to decode its transcriptional regulatory network (TRN), identifying 35 distinct signals that modulate specific gene sets. Our findings indicate that the ICA provides a qualitative advancement and captures nuanced relationships within gene clusters that other methods may miss. This study uncovers the fundamental properties of L. reuteri's TRN and deepens our understanding of its arginine metabolism and the co-regulation of riboflavin metabolism and fatty acid conversion. It also sheds light on conditions that regulate genes within a specific biosynthetic gene cluster and allows for the speculation of the potential role of isoprenoid biosynthesis in L. reuteri's adaptive response to environmental changes. By integrating transcriptomics and machine learning, we provide a system-level understanding of L. reuteri's response mechanism to environmental fluctuations, thus setting the stage for modeling the probiotic transcriptome for applications in microbial food production. IMPORTANCE We have studied Limosilactobacillus reuteri, a beneficial probiotic microbe that plays a significant role in our health and production of sustainable foods, a type of foods that are nutritionally dense and healthier and have low-carbon emissions compared to traditional foods. Similar to how humans adapt their lifestyles to different environments, this microbe adjusts its behavior by modulating the expression of genes. We applied machine learning to analyze large-scale data sets on how these genes behave across diverse conditions. From this, we identified 35 unique patterns demonstrating how L. reuteri adjusts its genes based on 50 unique environmental conditions (such as various sugars, salts, microbial cocultures, human milk, and fruit juice). This research helps us understand better how L. reuteri functions, especially in processes like breaking down certain nutrients and adapting to stressful changes. More importantly, with our findings, we become closer to using this knowledge to improve how we produce more sustainable and healthier foods with the help of microbes.
Collapse
Affiliation(s)
- Jonathan Josephs-Spaulding
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Copenhagen, Denmark
| | - Akanksha Rajput
- Department of Bioengineering, University of California, San Diego, California, USA
| | - Ying Hefner
- Department of Bioengineering, University of California, San Diego, California, USA
| | - Richard Szubin
- Department of Bioengineering, University of California, San Diego, California, USA
| | | | - Gaoyuan Li
- Department of Bioengineering, University of California, San Diego, California, USA
| | - Daniel C. Zielinski
- Department of Bioengineering, University of California, San Diego, California, USA
| | - Leonie Jahn
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Copenhagen, Denmark
| | - Morten Sommer
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Copenhagen, Denmark
| | - Patrick Phaneuf
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Copenhagen, Denmark
| | - Bernhard O. Palsson
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Copenhagen, Denmark
- Department of Bioengineering, University of California, San Diego, California, USA
| |
Collapse
|
11
|
Chang HY, Lin CY, Chiang Chiau JS, Chang JH, Hsu CH, Ko MHJ, Lee HC. Probiotic supplementation modifies the gut microbiota profile of very low birth weight preterm infants during hospitalization. Pediatr Neonatol 2024; 65:55-63. [PMID: 37500417 DOI: 10.1016/j.pedneo.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/09/2023] [Accepted: 06/07/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Probiotic supplementation is increasingly being given to very low birth weight (VLBW) preterm infants. This preliminary observational study aimed to investigate the effects of multiple-strain probiotics on the gut microbiota of VLBW preterm infants. METHODS We collected meconium and stool samples on days 14, 30, and 60 after birth from 49 VLBW infants with a gestational age of <32 weeks. The infants were divided into the probiotics (n = 24) and control (n = 25) groups. The microbial composition and diversity in the gut of the two groups were analyzed using 16 S rRNA gene sequencing. RESULTS The relative abundance of Bifidobacterium and Lactobacillus was significantly higher in the probiotics group than in the control group on days 14, 30, and 60 (Bifidobacterium: p = 0.002, p < 0.0001, and p < 0.0001, respectively; Lactobacillus: p = 0.012, p < 0.0001, and p < 0.0001, respectively). The control group exhibited a significantly higher proportion of participants with a low abundance (<1%) of Bifidobacterium or Lactobacillus on days 14, 30, and 60 than those in the probiotic group. Moreover, the probiotics group exhibited a significantly lower abundance of Klebsiella on days 14 and 30 (2.4% vs. 11.6%, p = 0.037; and 7.9% vs. 16.6%, p = 0.032, respectively) and of Escherichia-Shigella on day 60 than the control group (6.1% vs. 12.3%, p = 0.013). Beta diversity analysis revealed that the microbiota profile was clearly divided into two groups on days 30 and 60 (p = 0.001). CONCLUSION Probiotic supplementation significantly increased the relative abundance of Bifidobacterium and Lactobacillus and inhibited the growth of potential pathogens. Furthermore, probiotic supplementation led to a distinct gut microbiota profile. Further research is needed to identify probiotic strains that exert significant influence on the gut microbiome and their long-term health implications in preterm infants.
Collapse
Affiliation(s)
- Hung-Yang Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chia-Ying Lin
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan
| | | | - Jui-Hsing Chang
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chyong-Hsin Hsu
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan
| | - Mary Hsin-Ju Ko
- Department of Pediatrics, Hsinchu MacKay Memorial Hospital, Hsinchu 300, Taiwan
| | - Hung-Chang Lee
- Department of Pediatrics, MacKay Children's Hospital, Taipei 104, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan.
| |
Collapse
|
12
|
Batta VK, Rao SC, Patole SK. Bifidobacterium infantis as a probiotic in preterm infants: a systematic review and meta-analysis. Pediatr Res 2023; 94:1887-1905. [PMID: 37460707 PMCID: PMC10665187 DOI: 10.1038/s41390-023-02716-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/08/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Bifidobacterium infantis has special abilities to utilise human milk oligosaccharides. Hence we hypothesised that probiotic supplements containing B. infantis may confer greater benefits to preterm infants than probiotic supplements without B. infantis. METHODS A systematic review with meta-analysis was conducted according to standard guidelines. We selected RCTs evaluating probiotics compared to placebo or no treatment in preterm and/or low birth weight infants. Probiotic effects on Necrotizing Enterocolitis (NEC), Late Onset Sepsis (LOS) and Mortality were analysed separately for RCTs in which the supplemented probiotic product contained B. infantis and those that did not contain B. infantis. RESULTS 67 RCTs were included (n = 14,606), of which 16 used probiotics containing B. infantis (Subgroup A) and 51 RCTs did not (Subgroup B) Meta-analysis of all RCTs indicated that probiotics reduced the risk of NEC, LOS, and mortality. The subgroup meta-analysis demonstrated greater reduction in the incidence of NEC in subgroup A than subgroup B [(relative risk in subgroup A: 0.38; 95% CI, 0.27-0.55) versus (0.67; 95% CI, 0.55-0.81) in subgroup B; p value for subgroup difference: 0.01]. CONCLUSIONS These results provide indirect evidence that probiotic supplements that include B. infantis may be more beneficial for preterm infants. Well-designed RCTs are necessary to confirm these findings. IMPACT Evidence is emerging that beneficial effects of probiotics are species and strain specific. This systematic review analyses if B. infantis supplementation provides an advantage to preterm infants. This is the first systematic review evaluating the effects of probiotics containing B. infantis in preterm infants. The results of this systematic review provides indirect evidence that probiotics that include B. infantis may be more beneficial for preterm infants. These results will help in guiding future research and clinical practice for using B. infantis as a probiotic in preterm infants.
Collapse
Affiliation(s)
- Vamsi K Batta
- Neonatal Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia
- Neonatal Intensive Care Unit, King Edward Memorial Hospital, Perth, WA, Australia
| | - Shripada C Rao
- Neonatal Intensive Care Unit, Perth Children's Hospital, Perth, WA, Australia.
- School of Medicine, University of Western Australia, Perth, WA, Australia.
| | - Sanjay K Patole
- Neonatal Intensive Care Unit, King Edward Memorial Hospital, Perth, WA, Australia
- School of Medicine, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
13
|
Wala SJ, Sajankila N, Ragan MV, Duff AF, Wickham J, Volpe SG, Wang Y, Conces M, Dumbauld Z, Purayil N, Narayanan S, Rajab A, Mihi B, Bailey MT, Goodman SD, Besner GE. Superior performance of biofilm versus planktonic Limosilactobacillus reuteri in protection of the intestines and brain in a piglet model of necrotizing enterocolitis. Sci Rep 2023; 13:17740. [PMID: 37872187 PMCID: PMC10593788 DOI: 10.1038/s41598-023-44676-5] [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: 07/06/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of gastrointestinal-related death in premature infants. Its etiology is multifactorial, with intestinal dysbiosis playing a major role. Probiotics are a logical preventative therapy for NEC, however their benefits have been inconsistent. We previously developed a novel probiotic delivery system in which planktonic (free-living) Limosilactobacillus reuteri (Lr) is incubated with biocompatible dextranomer microspheres (DM) loaded with maltose (Lr-DM-maltose) to induce biofilm formation. Here we have investigated the effects of Lr-DM-maltose in an enteral feed-only piglet model of NEC. We found a significant decrease in the incidence of Definitive NEC (D-NEC), death associated with D-NEC, and activated microglia in the brains of piglets treated with Lr-DM-maltose compared to non-treated piglets. Microbiome analyses using 16S rRNA sequencing of colonic contents revealed a significantly different microbial community composition between piglets treated with Lr-DM-maltose compared to non-treated piglets, with an increase in Lactobacillaceae and a decrease in Clostridiaceae in Lr-DM-maltose-treated piglets. Furthermore, there was a significant decrease in the incidence of D-NEC between piglets treated with Lr-DM-maltose compared to planktonic Lr. These findings validate our previous results in rodents, and support future clinical trials of Lr in its biofilm state for the prevention of NEC in premature neonates.
Collapse
Affiliation(s)
- Samantha J Wala
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Nitin Sajankila
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Mecklin V Ragan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Audrey F Duff
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Joseph Wickham
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Samuel G Volpe
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Yijie Wang
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Miriam Conces
- Department of Pathology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zachary Dumbauld
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Nanditha Purayil
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Siddharth Narayanan
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Adrian Rajab
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Belgacem Mihi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michael T Bailey
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Steven D Goodman
- Center for Microbial Pathogenesis, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatric Surgery, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| |
Collapse
|
14
|
Rahkola EN, Rautava S, Hiltunen H, Ross C, Lahti L, Isolauri E. The preterm gut microbiota and administration routes of different probiotics: a randomized controlled trial. Pediatr Res 2023; 94:1480-1487. [PMID: 37020105 PMCID: PMC10589095 DOI: 10.1038/s41390-023-02560-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Preterm children with their aberrant gut microbiota and susceptibility to infections and inflammation constitute a considerable target group for probiotic therapy to generate the age-appropriate healthy microbiota. METHODS 68 preterm neonates were randomized into five intervention groups: Beginning from the median age of 3 days, 13 children received Lactobacillus rhamnosus GG (LGG) directly orally, and 17 via the lactating mother. 14 children received LGG with Bifidobacterium lactis Bb-12 (Bb12) orally, and 10 via the lactating mother. 14 children received placebo. The children's faecal microbiota was assessed at the age of 7 days by 16S rRNA gene sequencing. RESULTS The gut microbiota compositions of the children directly receiving the probiotic combination (LGG + Bb12) were significantly different from those of the children receiving the other intervention modes or placebo (p = 0.0012; PERMANOVA), the distinction being due to an increase in the relative abundance of Bifidobacterium animalis (P < 0.00010; ANCOM-BC), and the order Lactobacillales (P = 0.020; ANCOM-BC). CONCLUSION The connection between aberrant primary gut microbiota and a heightened risk of infectious and non-communicable diseases invites effective microbiota modulation. We show that the direct, early, and brief probiotic intervention of LGG + Bb12 109 CFU each, is sufficient to modulate the gut microbiota of the preterm neonate. IMPACT Preterm children have a higher risk of several health problems partly due to their aberrant gut microbiota. More research is needed to find a safe probiotic intervention to modify the gut microbiota of preterm children. The maternal administration route via breast milk might be safer for the newborn. In our study, the early and direct administration of the probiotic combination Lactobacillus rhamnosus GG with Bifidobacterium lactis Bb-12 increased the proportion of bifidobacteria in the preterm children's gut at the age of 7 days, but the maternal administration route was not as effective.
Collapse
Affiliation(s)
- Ella-Noora Rahkola
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland.
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland.
| | - Samuli Rautava
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Pediatrics, University of Helsinki and Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Henni Hiltunen
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Chandler Ross
- Department of Computing, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Erika Isolauri
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
15
|
Chen X, Shi Y. Determinants of microbial colonization in the premature gut. Mol Med 2023; 29:90. [PMID: 37407941 DOI: 10.1186/s10020-023-00689-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Abnormal microbial colonization in the gut at an early stage of life affects growth, development, and health, resulting in short- and long-term adverse effects. Microbial colonization patterns of preterm infants differ from those of full-term infants in that preterm babies and their mothers have more complicated prenatal and postnatal medical conditions. Maternal complications, antibiotic exposure, delivery mode, feeding type, and the use of probiotics may significantly shape the gut microbiota of preterm infants at an early stage of life; however, these influences subside with age. Although some factors and processes are difficult to intervene in or avoid, understanding the potential factors and determinants will help in developing timely strategies for a healthy gut microbiota in preterm infants. This review discusses potential determinants of gut microbial colonization in preterm infants and their underlying mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
16
|
Nakandalage R, Guan LL, Malmuthuge N. Microbial Interventions to Improve Neonatal Gut Health. Microorganisms 2023; 11:1328. [PMID: 37317302 DOI: 10.3390/microorganisms11051328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/04/2023] [Accepted: 05/16/2023] [Indexed: 06/16/2023] Open
Abstract
The diverse pioneer microbial community colonizing the mammalian gastrointestinal tract is critical for the developing immune system. Gut microbial communities of neonates can be affected by various internal and external factors, resulting in microbial dysbiosis. Microbial dysbiosis during early life affects gut homeostasis by changing metabolic, physiological, and immunological status, which increases susceptibility to neonatal infections and long-term pathologies. Early life is crucial for the establishment of microbiota and the development of the host immune system. Therefore, it provides a window of opportunity to reverse microbial dysbiosis with a positive impact on host health. Recent attempts to use microbial interventions during early life have successfully reversed dysbiotic gut microbial communities in neonates. However, interventions with persistent effects on microbiota and host health are still limited. This review will critically discuss microbial interventions, modulatory mechanisms, their limitations, and gaps in knowledge to understand their roles in improving neonatal gut health.
Collapse
Affiliation(s)
- Ranga Nakandalage
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Lethbridge Research and Development Center, Agriculture Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Nilusha Malmuthuge
- Lethbridge Research and Development Center, Agriculture Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| |
Collapse
|
17
|
Neumann CJ, Mahnert A, Kumpitsch C, Kiu R, Dalby MJ, Kujawska M, Madl T, Kurath-Koller S, Urlesberger B, Resch B, Hall LJ, Moissl-Eichinger C. Clinical NEC prevention practices drive different microbiome profiles and functional responses in the preterm intestine. Nat Commun 2023; 14:1349. [PMID: 36906612 PMCID: PMC10008552 DOI: 10.1038/s41467-023-36825-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/13/2023] [Indexed: 03/13/2023] Open
Abstract
Preterm infants with very low birthweight are at serious risk for necrotizing enterocolitis. To functionally analyse the principles of three successful preventive NEC regimens, we characterize fecal samples of 55 infants (<1500 g, n = 383, female = 22) longitudinally (two weeks) with respect to gut microbiome profiles (bacteria, archaea, fungi, viruses; targeted 16S rRNA gene sequencing and shotgun metagenomics), microbial function, virulence factors, antibiotic resistances and metabolic profiles, including human milk oligosaccharides (HMOs) and short-chain fatty acids (German Registry of Clinical Trials, No.: DRKS00009290). Regimens including probiotic Bifidobacterium longum subsp. infantis NCDO 2203 supplementation affect microbiome development globally, pointing toward the genomic potential to convert HMOs. Engraftment of NCDO 2203 is associated with a substantial reduction of microbiome-associated antibiotic resistance as compared to regimens using probiotic Lactobacillus rhamnosus LCR 35 or no supplementation. Crucially, the beneficial effects of Bifidobacterium longum subsp. infantis NCDO 2203 supplementation depends on simultaneous feeding with HMOs. We demonstrate that preventive regimens have the highest impact on development and maturation of the gastrointestinal microbiome, enabling the establishment of a resilient microbial ecosystem that reduces pathogenic threats in at-risk preterm infants.
Collapse
Affiliation(s)
- Charlotte J Neumann
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Alexander Mahnert
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Christina Kumpitsch
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria
| | - Raymond Kiu
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Matthew J Dalby
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Magdalena Kujawska
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health; Technical University of Munich, Freising, Bavaria, 85354, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology & Biochemistry, Medical University of Graz, Graz, Styria, 8010, Austria
- BioTechMed, Graz, Styria, 8010, Austria
| | - Stefan Kurath-Koller
- Division of Paediatric Cardiology, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria
| | - Berndt Urlesberger
- Division of Neonatology; Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Medical University of Graz, Graz, Styria, 8036, Austria
| | - Bernhard Resch
- Division of Neonatology; Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Graz, Styria, 8036, Austria.
- Research Unit for Neonatal Infectious Diseases and Epidemiology, Medical University of Graz, Graz, Styria, 8036, Austria.
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health; Technical University of Munich, Freising, Bavaria, 85354, Germany
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Christine Moissl-Eichinger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine; Medical University of Graz, Graz, Styria, 8010, Austria.
- BioTechMed, Graz, Styria, 8010, Austria.
| |
Collapse
|
18
|
Sajankila N, Wala SJ, Ragan MV, Volpe SG, Dumbauld Z, Purayil N, Mihi B, Besner GE. Current and future methods of probiotic therapy for necrotizing enterocolitis. Front Pediatr 2023; 11:1120459. [PMID: 36937955 PMCID: PMC10017871 DOI: 10.3389/fped.2023.1120459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a complex intestinal disease that primarily affects premature neonates. Given its significant mortality and morbidity, there is an urgent need to develop improved prophylactic measures against the disease. One potential preventative strategy for NEC is the use of probiotics. Although there has been significant interest for decades in probiotics in neonatal care, no clear guidelines exist regarding which probiotic to use or for which patients, and no FDA-approved products exist on the market for NEC. In addition, there is lack of agreement regarding the benefits of probiotics in neonates, as well as some concerns about the safety and efficacy of available products. We discuss currently available probiotics as well as next-generation probiotics and novel delivery strategies which may offer an avenue to capitalize on the benefits of probiotics, while minimizing the risks. Thus, probiotics may still prove to be an effective prevention strategy for NEC, although further product development and research is needed to support use in the preterm population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gail E. Besner
- Department of Pediatric Surgery, Center for Perinatal Research, Nationwide Children’s Hospital, Columbus, OH, United States
| |
Collapse
|
19
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
20
|
Neves LL, Hair AB, Preidis GA. A systematic review of associations between gut microbiota composition and growth failure in preterm neonates. Gut Microbes 2023; 15:2190301. [PMID: 36927287 PMCID: PMC10026866 DOI: 10.1080/19490976.2023.2190301] [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: 11/14/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Growth failure is among the most prevalent and devastating consequences of prematurity. Up to half of all extremely preterm neonates struggle to grow despite modern nutrition practices. Although elegant preclinical models suggest causal roles for the gut microbiome, these insights have not yet translated into biomarkers that identify at-risk neonates or therapies that prevent or treat growth failure. This systematic review aims to identify features of the neonatal gut microbiota that are positively or negatively associated with early postnatal growth. We identified 860 articles, of which 14 were eligible for inclusion. No two studies used the same definitions of growth, ages at stool collection, and statistical methods linking microbiota to metadata. In all, 58 different taxa were associated with growth, with little consensus among studies. Two or more studies reported positive associations with Enterobacteriaceae, Bacteroides, Bifidobacterium, Enterococcus, and Veillonella, and negative associations with Citrobacter, Klebsiella, and Staphylococcus. Streptococcus was positively associated with growth in five studies and negatively associated with growth in three studies. To gain insight into how the various definitions of growth could impact results, we performed an exploratory secondary analysis of 245 longitudinally sampled preterm infant stools, linking microbiota composition to multiple clinically relevant definitions of neonatal growth. Within this cohort, every definition of growth was associated with a different combination of microbiota features. Together, these results suggest that the lack of consensus in defining neonatal growth may limit our capacity to detect consistent, meaningful clinical associations that could be leveraged into improved care for preterm neonates.
Collapse
Affiliation(s)
- Larissa L. Neves
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Amy B. Hair
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
21
|
DeVeaux A, Ryou J, Dantas G, Warner BB, Tarr PI. Microbiome-targeting therapies in the neonatal intensive care unit: safety and efficacy. Gut Microbes 2023; 15:2221758. [PMID: 37358104 PMCID: PMC10294772 DOI: 10.1080/19490976.2023.2221758] [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: 12/06/2022] [Accepted: 05/25/2023] [Indexed: 06/27/2023] Open
Abstract
Microbiome-targeting therapies have received great attention as approaches to prevent disease in infants born preterm, but their safety and efficacy remain uncertain. Here we summarize the existing literature, focusing on recent meta-analyses and systematic reviews that evaluate the performance of probiotics, prebiotics, and/or synbiotics in clinical trials and studies, emphasizing interventions for which the primary or secondary outcomes were prevention of necrotizing enterocolitis, late-onset sepsis, feeding intolerance, and/or reduction in hospitalization length or all-cause mortality. Current evidence suggests that probiotics and prebiotics are largely safe but conclusions regarding their effectiveness in the neonatal intensive care unit have been mixed. To address this ambiguity, we evaluated publications that collectively support benefits of probiotics with moderate to high certainty evidence in a recent comprehensive network meta-analysis, highlighting limitations in these trials that make it difficult to support with confidence the routine, universal administration of probiotics to preterm infants.
Collapse
Affiliation(s)
- Anna DeVeaux
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian Ryou
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara B. Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Phillip I. Tarr
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
22
|
Mercer EM, Arrieta MC. Probiotics to improve the gut microbiome in premature infants: are we there yet? Gut Microbes 2023; 15:2201160. [PMID: 37122152 PMCID: PMC10153018 DOI: 10.1080/19490976.2023.2201160] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/06/2023] [Indexed: 05/02/2023] Open
Abstract
Gut microbiome maturation in infants born prematurely is uniquely influenced by the physiological, clinical, and environmental factors surrounding preterm birth and early life, leading to altered patterns of microbial succession relative to term infants during the first months of life. These differences in microbiome composition are implicated in acute clinical conditions that disproportionately affect preterm infants, including necrotizing enterocolitis (NEC) and late-onset sepsis (LOS). Probiotic supplementation initiated early in life is an effective prophylactic measure for preventing NEC, LOS, and other clinical concerns relevant to preterm infants. In parallel, reported benefits of probiotics on the preterm gut microbiome, metabolome, and immune function are beginning to emerge. This review summarizes the current literature on the influence of probiotics on the gut microbiome of preterm infants, outlines potential mechanisms by which these effects are exerted, and highlights important clinical considerations for determining the best practices for probiotic use in premature infants.
Collapse
Affiliation(s)
- Emily M. Mercer
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Wang G, Wang X, Ma Y, Cai S, Yang L, Fan Y, Zeng X, Qiao S. Lactobacillus reuteri improves the development and maturation of fecal microbiota in piglets through mother-to-infant microbe and metabolite vertical transmission. MICROBIOME 2022; 10:211. [PMID: 36461096 PMCID: PMC9717520 DOI: 10.1186/s40168-022-01336-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 07/26/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The immature neonatal fecal microbiota substantially impacts the development of gut health and greatly increases the risk of disease. Developing effective strategies to modulate the development of neonatal fecal microbiota has great significance. Herein, we investigated whether the maternal dietary supplementation and oral administration of Lactobacillus reuteri could effectively promote the development and maturation of the fecal microbiome in piglets from birth to weaning. RESULTS Metagenomic analysis of colostrum showed that maternal dietary L. reuteri supplementation influenced the overall microbiota composition, decreased the abundance of the phylum Proteobacteria and increased that of the species Bifidobacterium choerinum. KEGG pathway analysis revealed that maternal L. reuteri supplementation enriched the lysine biosynthesis and glycolysis/gluconeogenesis pathways and downregulated the bacterial invasion of epithelial cells in the colostrum. In addition, L. reuteri supplementation significantly altered the metabolite features and modules in umbilical cord blood serum based on metabolomics. Further, a significant covariation was observed between these differential metabolites and the species in colostrum. Maternal dietary L. reuteri supplementation also significantly influenced the microbiota composition and increased the meconium abundance of beneficial bacteria (such as Romboutsia, Lactobacillus, Blautia, Butyricicoccus, and Ruminococcus), some of which were markedly associated with several differential metabolites in umbilical cord blood serum between two groups. Notably, both the maternal dietary supplementation and oral intake of L. reuteri had strong impacts on the overall microbial composition and maturation of fecal microbiota in piglets during early life, and these effects were dependent on the growth stage. Oral administration of L. reuteri promoted diarrhea resistance in neonates, while maternal supplementation of L. reuteri enhanced the abilities of antioxidants and decreased inflammation. Moreover, the administration of L. reuteri via both methods in combination improved the growth performances of piglets. CONCLUSION Overall, our data demonstrated that L. reuteri had the ability to modulate the composition of fecal microbiota in newborn piglets by influencing the microbial community and functional composition in the colostrum and by altering several key metabolites in the umbilical cord blood serum. Also, both the maternal dietary supplementation and oral administration of L. reuteri effectively promoted the development and maturation of the fecal microbiome in piglets during early life. Both the maternal dietary supplementation and oral administration of L. reuteri in combination optimized the growth performances of piglets. Video Abstract.
Collapse
Affiliation(s)
- Gang Wang
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Xinyu Wang
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Yonghang Ma
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Shuang Cai
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Lijie Yang
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Yuxin Fan
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Xiangfang Zeng
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| | - Shiyan Qiao
- Present Address: State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
- Present Address: Beijing Key Laboratory of Biological Feed Additive, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
24
|
Chen X, Yan Z, Liu L, Zhang R, Zhang X, Peng C, Geng Y, Zhou F, Han Y, Hou X. Characteristics of gut microbiota of term small gestational age infants within 1 week and their relationship with neurodevelopment at 6 months. Front Microbiol 2022; 13:912968. [PMID: 36090083 PMCID: PMC9449527 DOI: 10.3389/fmicb.2022.912968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Small for gestational age (SGA) infants are at a higher risk of neurodevelopmental delay than infants appropriate for gestational age (AGA). Previous studies have confirmed that gut microbiota in early life influences subsequent neurodevelopment. However, few studies have reported corresponding data in SGA populations. Objective We aimed to evaluate the characteristics of the gut microbiota of term SGA infants and the associations between the gut microbiota in SGA infants and neurodevelopmental outcomes at 6 months of age. Methods Fecal samples were collected on days 1, 3, 5, and 7 from term SGA and AGA infants born between June 2020 and June 2021 at the Peking University First Hospital. 16S ribosomal deoxyribonucleic acid amplicon sequencing was used to analyze the fecal microbiota. We followed up for 6 months and used the Ages and Stages Questionnaires-3 (ASQ-3) to evaluate the neurodevelopmental outcomes among SGA infants. Results A total of 162 neonates were enrolled, with 41 SGA infants (25.3%) in the study group and 121 AGA infants (74.7%) in the control group. The gut microbial diversity in the SGA group was lower than that in the AGA group on days 1, 3, 5, and 7. Non-metric multidimensional scaling and analysis of similarities showed significant differences between the two groups. The SGA group had increased relative abundances of Ralstonia (3, 5, and 7 days) and Clostridium (3 and 7 days). The dominant microorganisms of the SGA group were Ralstonia on day 1, Escherichia_Shigella on days 3 and 7, and Clostridia on day 5. We found that the gut microbial diversity of SGA infants with poor communication scores was higher than that of SGA infants with good communication scores on day 3. Fine motor scores were negatively correlated with the relative abundance of Bacteroides_fragilis on day 1. A negative correlation was observed between gross motor scores and relative abundance of Clostridium_saccharobutylicum on day 7. Bacteroidota, Bacteroidia, Bacteroides, and Bacteroides_fragilis were the dominant microorganisms in the good communication score group on day 7. Communication scores were positively correlated with the relative abundance of Bacteroidota, Bacteroides, and Bacteroides_fragilis on day 7. Conclusion The gut microbial diversity of term SGA infants was significantly lower in the first week of life than that of term AGA infants. Certain pathogenic and conditional pathogenic bacteria, such as Escherichia_Shigella, Ralstonia and Clostridium increased or formed the dominant microbiota in SGA infants. Alpha diversity, Bacteroidota, Bacteroides, Bacteroides_fragilis, and Clostridium_saccharobutylicum found in SGA infants may be associated with neurodevelopmental outcomes at 6 months of age, indicating possible therapeutic targets for clinical intervention.
Collapse
|
25
|
Jia L, Wu J, Lei Y, Kong F, Zhang R, Sun J, Wang L, Li Z, Shi J, Wang Y, Wei Y, Zhang K, Lei Z. Oregano Essential Oils Mediated Intestinal Microbiota and Metabolites and Improved Growth Performance and Intestinal Barrier Function in Sheep. Front Immunol 2022; 13:908015. [PMID: 35903106 PMCID: PMC9314563 DOI: 10.3389/fimmu.2022.908015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
With the increased demand for safe and sustainable alternatives to growth promoting antibiotics in the livestock industry, oregano essential oils (OEO) and Lactobacillus reuteri (LR) have been examined as alternatives to antibiotics for growth promotion and to improve animal health and performance. However, the mechanism underlying the OEO and LR mediation of sheep growth remains unknown. In this study, 16S rRNA gene sequencing and untargeted metabolomics were used to determine the role of the gut microbiota in the growth improvements observed. The potential modulating roles of intestinal microbial metabolites of OEO and LR to intestinal health were systematically explored as well. It was observed that both OEO and LR had greater average daily gain (ADG) and lower F/G ratio. Furthermore, OEO also appeared to have produced a greater amylase enzyme activity and mucin gene expression in the jejunal mucosa. It was also observed that OEO reduced serum IL-2 and TNF-β as well as mRNA levels of NF-κB p65, toll-like receptor-4 (TLR-4), and IL-6 in the jejunal mucosa. Moreover, dietary OEO supplementation increased the abundances of Ruminococcus, Bifidobacterium and Enterococcus, while the relative abundances of Succiniclasticum, Marvinbryantia and Streptococcus were enriched in LR group. Spearman’s correlation analysis revealed that the abundances of Bifidobacterium, Ruminococcus and Enterococcus were positively correlated with the mRNA expression of mucins. Moreover, the relative abundance of Enterococcus was positively correlated with amylase activity. Metabolomics analysis indicated that OEO and LR increased the levels of indole acetaldehyde and indole-3-acetic acid through the tryptophan metabolism pathway. It was observed that LR also decreased the inflammatory metabolites including tryptamine and 5-hydroxyindole-3-acetic acid. Collectively, these results suggested that OEO exerted a beneficial effect on growth performance and the mucosal barrier, affected tryptophan metabolism and improved the intestinal microbiota of sheep.
Collapse
Affiliation(s)
- Li Jia
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jianping Wu
- Institute of Rural Development, Northwest Normal University, Lanzhou, China
| | - Yu Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Fanyun Kong
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Rui Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jianxiang Sun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Liao Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zemin Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jinping Shi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Ying Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yubing Wei
- The Animal Husbandry and Veterinary Station in Pingshan Lake Mongolian Township of Ganzhou District, Zhangye, China
| | - Ke Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhaomin Lei
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- *Correspondence: Zhaomin Lei,
| |
Collapse
|
26
|
Horne RG, Freedman SB, Johnson-Henry KC, Pang XL, Lee BE, Farion KJ, Gouin S, Schuh S, Poonai N, Hurley KF, Finkelstein Y, Xie J, Williamson-Urquhart S, Chui L, Rossi L, Surette MG, Sherman PM. Intestinal Microbial Composition of Children in a Randomized Controlled Trial of Probiotics to Treat Acute Gastroenteritis. Front Cell Infect Microbiol 2022; 12:883163. [PMID: 35774405 PMCID: PMC9238408 DOI: 10.3389/fcimb.2022.883163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Compositional analysis of the intestinal microbiome in pre-schoolers is understudied. Effects of probiotics on the gut microbiota were evaluated in children under 4-years-old presenting to an emergency department with acute gastroenteritis. Included were 70 study participants (n=32 placebo, n=38 probiotics) with stool specimens at baseline (day 0), day 5, and after a washout period (day 28). Microbiota composition and deduced functions were profiled using 16S ribosomal RNA sequencing and predictive metagenomics, respectively. Probiotics were detected at day 5 of administration but otherwise had no discernable effects, whereas detection of bacterial infection (P<0.001) and participant age (P<0.001) had the largest effects on microbiota composition, microbial diversity, and deduced bacterial functions. Participants under 1 year had lower bacterial diversity than older aged pre-schoolers; compositional changes of individual bacterial taxa were associated with maturation of the gut microbiota. Advances in age were associated with differences in gut microbiota composition and deduced microbial functions, which have the potential to impact health later in life.
Collapse
Affiliation(s)
- Rachael G. Horne
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen B. Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Department of Pediatrics, Alberta Children’s Hospital, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Xiao-Li Pang
- Alberta Precision Laboratories – Public Health Laboratory (ProvLab), Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Bonita E. Lee
- Women and Children’s Research Institute, Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada
| | - Ken J. Farion
- Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| | - Serge Gouin
- Departments of Emergency Medicine and Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Suzanne Schuh
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Naveen Poonai
- Division of Pediatric Emergency Medicine, London Children’s Hospital Health Science Centre, Department of Pediatrics, Western University, London, ON, Canada
| | - Katrina F. Hurley
- Pediatric Emergency Medicine, Izaak Walton Killam (IWK) Children’s Hospital, Dalhousie University, Halifax, NS, Canada
| | - Yaron Finkelstein
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jianling Xie
- Pediatric Emergency Medicine, Izaak Walton Killam (IWK) Children’s Hospital, Dalhousie University, Halifax, NS, Canada
| | - Sarah Williamson-Urquhart
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children’s Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Linda Chui
- Alberta Precision Laboratories – Public Health Laboratory (ProvLab), Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Laura Rossi
- Department of Biochemistry and Biomedical Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Michael G. Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Philip M. Sherman
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- *Correspondence: Philip M. Sherman,
| |
Collapse
|
27
|
Athalye-Jape G, Esvaran M, Patole S, Simmer K, Nathan E, Doherty D, Keil A, Rao S, Chen L, Chandrasekaran L, Kok C, Schuster S, Conway P. Effect of single versus multistrain probiotic in extremely preterm infants: a randomised trial. BMJ Open Gastroenterol 2022; 9:bmjgast-2021-000811. [PMID: 35185013 PMCID: PMC8860036 DOI: 10.1136/bmjgast-2021-000811] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/12/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE Evidence indicates that multistrain probiotics benefit preterm infants more than single-strain (SS) probiotics. We assessed the effects of SS versus triple-strain (TS) probiotic supplementation (PS) in extremely preterm (EP) infants. DESIGN EP infants (gestational age (GA) <28 weeks) were randomly allocated to TS or SS probiotic, assuring blinding. Reference (REF) group was EP infants in the placebo arm of our previous probiotic trial. PS was commenced with feeds and continued until 37 weeks' corrected GA. Primary outcome was time to full feed (TFF: 150 mL/kg/day). Secondary outcomes included short-chain fatty acids and faecal microbiota collected at T1 (first week) and T2 (after 3 weeks of PS) using 16S ribosomal RNA gene sequencing. RESULTS 173 EP (SS: 86, TS: 87) neonates with similar GA and birth weight (BW) were randomised. Median TFF was comparable (11 (IQR 8-16) vs 10 (IQR 8-16) days, p=0.92). Faecal propionate (SS, p<0.001, and TS, p=0.0009) and butyrate levels (TS, p=0.029) were significantly raised in T2 versus T1 samples. Secondary clinical outcomes were comparable. At T2, alpha diversity was comparable (p>0.05) between groups, whereas beta-diversity analysis revealed significant differences between PS and REF groups (both p=0.001). Actinobacteria were higher (both p<0.01), and Proteobacteria, Firmicutes and Bacteroidetes were lower in PS versus REF. Gammaproteobacteria, Clostridia and Negativicutes were lower in both PS versus REF. CONCLUSION TFF in EP infants was similar between SS and TS probiotics. Both probiotics were effective in reducing dysbiosis (higher bifidobacteria and lower Gammaproteobacteria). Long-term significance of increased propionate and butyrate needs further studies. TRIAL REGISTRATION NUMBER ACTRN 12615000940572.
Collapse
Affiliation(s)
- Gayatri Athalye-Jape
- Neonatology directorate, King Edward Memorial Hospital for Women Perth, Subiaco, Western Australia, Australia
| | - Meera Esvaran
- Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Patole
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Karen Simmer
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Elizabeth Nathan
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Dorota Doherty
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Anthony Keil
- Microbiology, PathWest Laboratory Medicine Western Australia, Nedlands, Western Australia, Australia
| | - Shripada Rao
- Neonatal Clinical Care Unit, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Liwei Chen
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | | | - Chooi Kok
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Stephan Schuster
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | - Patricia Conway
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| |
Collapse
|
28
|
Westaway JAF, Huerlimann R, Kandasamy Y, Miller CM, Norton R, Watson D, Infante-Vilamil S, Rudd D. To Probiotic or Not to Probiotic: A Metagenomic Comparison of the Discharge Gut Microbiome of Infants Supplemented With Probiotics in NICU and Those Who Are Not. Front Pediatr 2022; 10:838559. [PMID: 35345612 PMCID: PMC8957066 DOI: 10.3389/fped.2022.838559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/09/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Preterm birth is associated with the development of both acute and chronic disease, and the disruption of normal gut microbiome development. Recent studies have sought to both characterize and understand the links between disease and the microbiome. Probiotic treatment may correct for these microbial imbalances and, in turn, mitigate disease. However, the criteria for probiotic supplementation in NICU's in North Queensland, Australia limits its usage to the most premature (<32 weeks gestation) and small for gestational age infants (<1,500 g). Here we use a combination of amplicon and shotgun metagenomic sequencing to compare the gut microbiome of infants who fulfill the criteria for probiotic-treatment and those who do not. The aims of this study were to determine if probiotic-supplemented preterm infants have significantly different taxonomic and functional profiles when compared to non-supplemented preterm infants at discharge. METHODS Preterm infants were recruited in North Queensland, Australia, with fecal samples collected just prior to discharge (36 ± 0.5 weeks gestation), to capture potential changes that could be probiotic induced. All samples underwent 16S rRNA gene amplicon sequencing, with a subset also used for shotgun metagenomics. Mixed effects models were used to assess the effect of probiotics on alpha diversity, beta diversity and taxonomic abundance, whilst accounting for other known covariates. RESULTS Mixed effects modeling demonstrated that probiotic treatment had a significant effect on overall community composition (beta diversity), characterized by greater alpha diversity and differing abundances of several taxa, including Bifidobacterium and Lactobacillus, in supplemented infants. CONCLUSION Late preterm-infants who go without probiotic-supplementation may be missing out on stabilizing-effects provided through increased alpha diversity and the presence of commensal microbes, via the use of probiotic-treatment. These findings suggest that late-preterm infants may benefit from probiotic supplementation. More research is needed to both understand the consequences of the differences observed and the long-term effects of this probiotic-treatment.
Collapse
Affiliation(s)
- Jacob A F Westaway
- College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia
| | - Roger Huerlimann
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia.,Marine Climate Change Unit, Okinawa Institute of Science and Technology (OIST), Onna, Japan.,Center for Sustainable Tropical Fisheries and Aquaculture, James Cook University, Townsville, QLD, Australia
| | - Yoga Kandasamy
- College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, QLD, Australia.,Neonatology, Townsville University Hospital, Townsville, QLD, Australia
| | - Catherine M Miller
- College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, QLD, Australia
| | - Robert Norton
- Microbiology, Pathology Queensland, Herston, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - David Watson
- Maternal-Fetal Medicine, Townsville University Hospital, Townsville, QLD, Australia
| | - Sandra Infante-Vilamil
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Townsville, QLD, Australia.,Center for Sustainable Tropical Fisheries and Aquaculture, James Cook University, Townsville, QLD, Australia
| | - Donna Rudd
- College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
29
|
Lee H, Li Z, Christensen B, Peng Y, Li X, Hernell O, Lönnerdal B, Slupsky CM. Metabolic Phenotype and Microbiome of Infants Fed Formula Containing Lactobacillus paracasei Strain F-19. Front Pediatr 2022; 10:856951. [PMID: 35558362 PMCID: PMC9087039 DOI: 10.3389/fped.2022.856951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Early childhood nutrition drives the development of the gut microbiota. In contrast to breastfeeding, feeding infant formula has been shown to impact both the gut microbiota and the serum metabolome toward a more unfavorable state. It is thought that probiotics may alter the gut microbiota and hence create a more favorable metabolic outcome. To investigate the impact of supplementation with Lactobacillus paracasei spp. paracasei strain F-19 on the intestinal microbiota and the serum metabolome, infants were fed a formula containing L. paracasei F19 (F19) and compared to a cohort of infants fed the same standard formula without the probiotic (SF) and a breast-fed reference group (BF). The microbiome, as well as serum metabolome, were compared amongst groups. Consumption of L. paracasei F19 resulted in lower community diversity of the gut microbiome relative to the SF group that made it more similar to the BF group at the end of the intervention (4 months). It also significantly increased lactobacilli and tended to increase bifidobacteria, also making it more similar to the BF group. The dominant genus in the microbiome of all infants was Bifidobacterium throughout the intervention, which was maintained at 12 months. Although the serum metabolome of the F19 group was more similar to the group receiving the SF than the BF group, increases in serum TCA cycle intermediates and decreases in several amino acids in the metabolome of the F19 group were observed, which resulted in a metabolome that trended toward the BF group. Overall, L. paracasei F19 supplementation did not override the impact of formula-feeding but did impact the microbiome and the serum metabolome in a way that may mitigate some unfavorable metabolic impacts of formula-feeding.
Collapse
Affiliation(s)
- Hanna Lee
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | | | - Yongmei Peng
- Department of Child Health Care, Children's Hospital, Fudan University, Shanghai, China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
30
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
31
|
Pediococcus pentosaceus IM96 Exerts Protective Effects against Enterohemorrhagic Escherichia coli O157:H7 Infection In Vivo. Foods 2021; 10:foods10122945. [PMID: 34945495 PMCID: PMC8700651 DOI: 10.3390/foods10122945] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a notorious and prevalent foodborne pathogen which can cause serious intestinal diseases. The antagonistic activity of probiotics against EHEC is promising, but most of the studies concerning this subject have been carried out in vitro. Specifically, the interaction between Pediococcus pentosaceus and EHEC O157:H7 in vivo has not been reported yet. In this study, we investigated the protective effect of P. pentosaceus IM96 on EHEC O157:H7-infected female mice in vivo. The results demonstrated that P. pentosaceus IM96 reduced the level of pro-inflammatory factors and increased the level of anti-inflammatory factors of EHEC O157:H7-infected mice. Furthermore, P. pentosaceus IM96 alleviated intestinal mucosal damage and increased the level of MUC-2, tight junction (TJ) proteins, and short chain fatty acids (SCFAs). The intestinal microbial community structure and the diversity and richness of the microbiota were also changed by P. pentosaceus IM96 treatment. In summary, P. pentosaceus IM96 exerted protective effects against EHEC O157:H7 via alleviating intestinal inflammation, strengthening the intestinal barrier function, and regulating intestinal microbiota, suggesting that P. pentosaceus IM96 might serve as a potential microbial agent to prevent and treat intestinal diseases caused by EHEC O157:H7 infection in the future.
Collapse
|
32
|
Hung YP, Lee CC, Lee JC, Tsai PJ, Hsueh PR, Ko WC. The Potential of Probiotics to Eradicate Gut Carriage of Pathogenic or Antimicrobial-Resistant Enterobacterales. Antibiotics (Basel) 2021; 10:antibiotics10091086. [PMID: 34572668 PMCID: PMC8470257 DOI: 10.3390/antibiotics10091086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/31/2022] Open
Abstract
Probiotic supplements have been used to decrease the gut carriage of antimicrobial-resistant Enterobacterales through changes in the microbiota and metabolomes, nutrition competition, and the secretion of antimicrobial proteins. Many probiotics have shown Enterobacterales-inhibiting effects ex vivo and in vivo. In livestock, probiotics have been widely used to eradicate colon or environmental antimicrobial-resistant Enterobacterales colonization with promising efficacy for many years by oral supplementation, in ovo use, or as environmental disinfectants. In humans, probiotics have been used as oral supplements for infants to decease potential gut pathogenic Enterobacterales, and probiotic mixtures, especially, have exhibited positive results. In contrast to the beneficial effects in infants, for adults, probiotic supplements might decrease potentially pathogenic Enterobacterales, but they fail to completely eradicate them in the gut. However, there are several ways to improve the effects of probiotics, including the discovery of probiotics with gut-protection ability and antimicrobial effects, the modification of delivery methods, and the discovery of engineered probiotics. The search for multifunctional probiotics and synbiotics could render the eradication of “bad” Enterobacterales in the human gut via probiotic administration achievable in the future.
Collapse
Affiliation(s)
- Yuan-Pin Hung
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan 700, Taiwan;
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Ching-Chi Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Clinical Medicine Research Center, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
| | - Jen-Chieh Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, School of Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (P.-R.H.); (W.-C.K.)
| | - Wen-Chien Ko
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Correspondence: (P.-R.H.); (W.-C.K.)
| |
Collapse
|
33
|
Martí M, Spreckels JE, Jenmalm MC, Abrahamsson T. A protocol for characterization of extremely preterm infant gut microbiota in double-blind clinical trials. STAR Protoc 2021; 2:100652. [PMID: 34308378 PMCID: PMC8283139 DOI: 10.1016/j.xpro.2021.100652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
16S rRNA gene sequencing enables microbial community profiling, but recovering fecal DNA from extremely premature infants is challenging. Here, we describe an optimized protocol for fecal DNA isolation, library preparation for 16S rRNA gene sequencing, taxonomy assignation, and statistical analyses. The protocol is complemented with a quantitative PCR for probiotic L. reuteri identification. This protocol describes how to characterize preterm infant gut microbiota and relate it to probiotic supplementation and clinical outcomes. It is customizable for other clinical trials. For complete details on the use and execution of this protocol, please refer to Martí et al. (2021) and Spreckels et al. (2021). Customizable protocol for characterization of infant gut microbiota in clinical trials Optimized sample preparation for 16S rRNA gene sequencing for infant fecal samples Quantitative PCR for probiotic Lactobacillus reuteri quantification in infant feces Easy-to-use bioinformatic and statistical analysis pipeline
Collapse
Affiliation(s)
- Magalí Martí
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanne E Spreckels
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Genetics, University Medical Centre Groningen, Groningen, the Netherlands
| | - Maria C Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Thomas Abrahamsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Paediatrics, Linköping University, Linköping, Sweden
| |
Collapse
|
34
|
Lactobacillus reuteri Colonisation of Extremely Preterm Infants in a Randomised Placebo-Controlled Trial. Microorganisms 2021; 9:microorganisms9050915. [PMID: 33923278 PMCID: PMC8190634 DOI: 10.3390/microorganisms9050915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022] Open
Abstract
Lactobacillus reuteri DSM 17938 supplementation reduces morbidities in very low birth weight infants (<1500 g), while the effect on extremely low birth weight infants (ELBW, <1000 g) is still questioned. In a randomised placebo-controlled trial (ClinicalTrials.gov ID NCT01603368), head growth, but not feeding tolerance or morbidities, improved in L. reuteri-supplemented preterm ELBW infants. Here, we investigate colonisation with the probiotic strain in preterm ELBW infants who received L. reuteri DSM 17938 or a placebo from birth to postmenstrual week (PMW) 36. Quantitative PCR was used on 582 faecal DNA samples collected from 132 ELBW infants at one, two, three, and four weeks, at PMW 36, and at two years of age. Human milk oligosaccharides were measured in 31 milk samples at two weeks postpartum. At least 86% of the ELBW infants in the L. reuteri group were colonised with the probiotic strain during the neonatal period, despite low gestational age, high antibiotic pressure, and independent of infant feeding mode. Higher concentrations of lacto-N-tetraose, sialyl-lacto-N-neotetraose c, and 6′-sialyllactose in mother’s milk weakly correlated with lower L. reuteri abundance. Within the L. reuteri group, higher L. reuteri abundance weakly correlated with a shorter time to reach full enteral feeding. Female sex and L. reuteri colonisation improved head growth from birth to four weeks of age. In conclusion, L. reuteri DSM 17938 supplementation leads to successful colonisation in ELBW infants.
Collapse
|