1
|
Nunez H, Nieto PA, Mars RA, Ghavami M, Sew Hoy C, Sukhum K. Early life gut microbiome and its impact on childhood health and chronic conditions. Gut Microbes 2025; 17:2463567. [PMID: 39916516 PMCID: PMC11810090 DOI: 10.1080/19490976.2025.2463567] [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: 09/23/2024] [Revised: 12/20/2024] [Accepted: 02/02/2025] [Indexed: 02/12/2025] Open
Abstract
The development of the gut microbiome is crucial to human health, particularly during the first three years of life. Given its role in immune development, disturbances in the establishment process of the gut microbiome may have long term consequences. This review summarizes evidence for these claims, highlighting compositional changes of the gut microbiome during this critical period of life as well as factors that affect gut microbiome development. Based on human and animal data, we conclude that the early-life microbiome is a determinant of long-term health, impacting physiological, metabolic, and immune processes. The early-life gut microbiome field faces challenges. Some of these challenges are technical, such as lack of standardized stool collection protocols, inconsistent DNA extraction methods, and outdated sequencing technologies. Other challenges are methodological: small sample sizes, lack of longitudinal studies, and poor control of confounding variables. To address these limitations, we advocate for more robust research methodologies to better understand the microbiome's role in health and disease. Improved methods will lead to more reliable microbiome studies and a deeper understanding of its impact on health outcomes.
Collapse
Affiliation(s)
- Harold Nunez
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
| | | | - Ruben A. Mars
- Seeding Inc, DBA Tiny Health, Austin, Texas, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
2
|
Klaassens ES, Baak ML, Mekkes NJ, Bongoni R, Schaubeck M. Effect of protein modification in synbiotic infant formula on probiotic metabolic activity and bacterial composition in an infant gut-model. MICROBIOME RESEARCH REPORTS 2024; 3:38. [PMID: 39421252 PMCID: PMC11480727 DOI: 10.20517/mrr.2024.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/30/2024] [Accepted: 06/25/2024] [Indexed: 10/19/2024]
Abstract
Aim: Microbial colonization of the neonatal gut is pivotal in priming the infant's immune system. Human milk (HM) is the best nutrition for infants and supports the development of the microbiota due to prebiotic compounds and probiotic microorganisms. When exclusive breastfeeding is not possible, infant formula (IF) with probiotics is a strategy to support the infant's microbiome development. However, knowledge about the effects of the infant gut microbiota and different compounds in IF on individual probiotic strains is limited, as strain-level detection in a complex ecosystem is challenging. The aim of the present study was to show the effects of IF with different protein forms on the metabolic activity of two probiotic strains isolated from HM in a complex ecosystem. Methods: By using an ex-vivo infant gut model containing infant donor-microbiota, the effects of IF with either intact or extensively hydrolyzed protein on the metabolic activity of the donor microbiota, as well as two probiotic strains [Limosilactobacillus fermentum (L. fermentum) CECT 5716 (Lf) and Bifidobacterium breve (B. breve) DSM 32583 (Bb)], were analyzed. A new bioinformatic pipeline combined with a specific infant microbiome database was used to explore shotgun metagenome datasets (1200 Megabases) for taxonomic identification and strain-level tracking. Results: Both protein forms (i.e., intact or extensively hydrolyzed protein) in IF supported infant gut microbial metabolic activity equally, as evidenced by similar levels of short-chain fatty acids (SCFAs). Interestingly, gut microbial metabolic activity was found to be differently activated in a strain-dependent manner. Taxonomic profiling of the microbiome at the strain level enabled monitoring of the prevalence and abundance of both probiotic strains, even in a complex ecosystem. Conclusion: Food matrix and host microbiota interactions should be considered when evaluating strain-specific probiotic effects in the future.
Collapse
Affiliation(s)
| | | | | | | | - Monika Schaubeck
- Research & Development, HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen 85276, Germany
| |
Collapse
|
3
|
Xu J, Duar RM, Quah B, Gong M, Tin F, Chan P, Sim CK, Tan KH, Chong YS, Gluckman PD, Frese SA, Kyle D, Karnani N. Delayed colonization of Bifidobacterium spp. and low prevalence of B. infantis among infants of Asian ancestry born in Singapore: insights from the GUSTO cohort study. Front Pediatr 2024; 12:1421051. [PMID: 38915873 PMCID: PMC11194334 DOI: 10.3389/fped.2024.1421051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Background The loss of ancestral microbes, or the "disappearing microbiota hypothesis" has been proposed to play a critical role in the rise of inflammatory and immune diseases in developed nations. The effect of this loss is most consequential during early-life, as initial colonizers of the newborn gut contribute significantly to the development of the immune system. Methods In this longitudinal study (day 3, week 3, and month 3 post-birth) of infants of Asian ancestry born in Singapore, we studied how generational immigration status and common perinatal factors affect bifidobacteria and Bifidobacterium longum subsp. infantis (B. infantis) colonization. Cohort registry identifier: NCT01174875. Results Our findings show that first-generation migratory status, perinatal antibiotics usage, and cesarean section birth, significantly influenced the abundance and acquisition of bifidobacteria in the infant gut. Most importantly, 95.6% of the infants surveyed in this study had undetectable B. infantis, an early and beneficial colonizer of infant gut due to its ability to metabolize the wide variety of human milk oligosaccharides present in breastmilk and its ability to shape the development of a healthy immune system. A comparative analysis of B. infantis in 12 countries by their GDP per capita showed a remarkably low prevalence of this microbe in advanced economies, especially Singapore. Conclusion This study provides new insights into infant gut microbiota colonization, showing the impact of generational immigration on early-life gut microbiota acquisition. It also warrants the need to closely monitor the declining prevalence of beneficial microbes such as B. infantis in developed nations and its potential link to increasing autoimmune and allergic diseases.
Collapse
Affiliation(s)
- Jia Xu
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | | | - Baoling Quah
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Min Gong
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Felicia Tin
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Penny Chan
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Clinical Data Engagement, Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Choon Kiat Sim
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Kok Hian Tan
- SingHealth Duke-NUS Institute for Patient Safety and Quality, Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Yap Seng Chong
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynecology and Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D. Gluckman
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Centre for SPDS Centre for Informed Futures, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Steven A. Frese
- Department of Nutrition, University of Nevada, Reno, NV, United States
| | - David Kyle
- Infinant Health, Inc., Davis, CA, United States
| | - Neerja Karnani
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Clinical Data Engagement, Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
4
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
5
|
Guo H, Cui J, Li Q, Liang X, Li J, Yang B, Kalds P, Chen Y, Yang Y. A multi-omic assessment of the mechanisms of intestinal microbes used to treat diarrhea in early-weaned lambs. mSystems 2024; 9:e0095323. [PMID: 38193712 PMCID: PMC10878098 DOI: 10.1128/msystems.00953-23] [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: 09/07/2023] [Accepted: 10/07/2023] [Indexed: 01/10/2024] Open
Abstract
Transplant of donor microbiota can significantly alter the structure of the host's intestinal microbiota and alleviate early weaning stress. Screening for alternative-resistant products by transplanting fecal bacteria from healthy lambs is a current research trend in the livestock industry. In the present study, fecal microbiota transplantation was performed in lambs with diarrhea during early weaning. The transplanted fecal microbiota greatly reduced the diarrhea and serum inflammatory factor levels caused by early weaning. Transcriptome sequencing revealed that fecal microbiota transplantation alleviated colonic inflammation and increased the expression of colonic ion transport proteins. In addition, the levels of Streptococcus, Enterococcus, and Escherichia Shigella decreased in the jejunum, cecum, and colon of the lambs; meanwhile, the levels of Bifidobacterium and multiple secondary bile acids, such as ursodeoxycholic acid, increased in the colon. Furthermore, the abundance of Bifidobacterium was significantly negatively correlated with the diarrhea index. The fecal microbiota transplantation reshaped the intestinal microbiota of early-weaned lambs, protected the intestinal physiology and immune barrier, and reduced weaning stress. In addition to making available bacteriological products for controlling intestinal inflammation in young lambs, this study offers a theoretical framework and technical system for the mechanisms by which microbiota transplantation regulates intestinal health in young lambs.IMPORTANCEBefore weaning, the digestive system of lambs is not well developed; hence, its resistance to infectious diseases is weak. Under intensive feeding systems, lambs can easily be stressed and the risk of bacterial infection is high, which causes diarrhea, which in turn may cause mortality and significant economic losses to the livestock industry. With the elimination of antibiotics in animal feed, the incidence of mortality due to intestinal illnesses in lambs has gradually increased. There are several types of probiotics routinely used in young animals, but the effects and processes of their usage have only been assessed in monogastric animals. The lack of data on ruminants, particularly sheep, has severely hampered the process of efficient and healthy sheep breeding. Therefore, there is an urgent need to identify effective and safe functional supplements for lambs.
Collapse
Affiliation(s)
- Hongran Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Jiuzeng Cui
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Qian Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Xuhui Liang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Junda Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Bohua Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Peter Kalds
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yuxin Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, China
| |
Collapse
|
6
|
Gavzy SJ, Kensiski A, Lee ZL, Mongodin EF, Ma B, Bromberg JS. Bifidobacterium mechanisms of immune modulation and tolerance. Gut Microbes 2023; 15:2291164. [PMID: 38055306 PMCID: PMC10730214 DOI: 10.1080/19490976.2023.2291164] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
Bifidobacterium is a widely distributed commensal bacterial genus that displays beneficial pro-homeostatic and anti-inflammatory immunomodulatory properties. Depletion or absence of Bifidobacterium in humans and model organisms is associated with autoimmune responses and impaired immune homeostasis. At the cellular level, Bifidobacterium upregulates suppressive regulatory T cells, maintains intestinal barrier function, modulates dendritic cell and macrophage activity, and dampens intestinal Th2 and Th17 programs. While there has been a large volume of literature characterizing the probiotic properties of various Bifidobacterial species, the likely multifactorial mechanisms underlying these effects remain elusive, in particular, its immune tolerogenic effect. However, recent work has shed light on Bifidobacterium surface structural polysaccharide and protein elements, as well as its metabolic products, as commensal mediators of immune homeostasis. This review aims to discuss several mechanisms Bifidobacterium utilizes for immune modulation as well as their indirect impact on the regulation of gut microbiome structure and function, from structural molecules to produced metabolites. These mechanisms are pertinent to an increasingly networked understanding of immune tolerance and homeostasis in health and disease.
Collapse
Affiliation(s)
- Samuel J Gavzy
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Allison Kensiski
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zachariah L Lee
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emmanuel F Mongodin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jonathan S Bromberg
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Button JE, Cosetta CM, Reens AL, Brooker SL, Rowan-Nash AD, Lavin RC, Saur R, Zheng S, Autran CA, Lee ML, Sun AK, Alousi AM, Peterson CB, Koh AY, Rechtman DJ, Jenq RR, McKenzie GJ. Precision modulation of dysbiotic adult microbiomes with a human-milk-derived synbiotic reshapes gut microbial composition and metabolites. Cell Host Microbe 2023; 31:1523-1538.e10. [PMID: 37657443 DOI: 10.1016/j.chom.2023.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/13/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
Manipulation of the gut microbiome using live biotherapeutic products shows promise for clinical applications but remains challenging to achieve. Here, we induced dysbiosis in 56 healthy volunteers using antibiotics to test a synbiotic comprising the infant gut microbe, Bifidobacterium longum subspecies infantis (B. infantis), and human milk oligosaccharides (HMOs). B. infantis engrafted in 76% of subjects in an HMO-dependent manner, reaching a relative abundance of up to 81%. Changes in microbiome composition and gut metabolites reflect altered recovery of engrafted subjects compared with controls. Engraftment associates with increases in lactate-consuming Veillonella, faster acetate recovery, and changes in indolelactate and p-cresol sulfate, metabolites that impact host inflammatory status. Furthermore, Veillonella co-cultured in vitro and in vivo with B. infantis and HMO converts lactate produced by B. infantis to propionate, an important mediator of host physiology. These results suggest that the synbiotic reproducibly and predictably modulates recovery of a dysbiotic microbiome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Martin L Lee
- Prolacta Bioscience, Duarte, CA 91010, USA; Department of Biostatistics, University of California Los Angeles, Fielding School of Public Health, Los Angeles, CA 90095, USA
| | - Adam K Sun
- Prolacta Bioscience, Duarte, CA 91010, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Robert R Jenq
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
8
|
Horrocks V, King OG, Yip AYG, Marques IM, McDonald JAK. Role of the gut microbiota in nutrient competition and protection against intestinal pathogen colonization. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001377. [PMID: 37540126 PMCID: PMC10482380 DOI: 10.1099/mic.0.001377] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023]
Abstract
The human gut microbiota can restrict the growth of pathogens to prevent them from colonizing the intestine ('colonization resistance'). However, antibiotic treatment can kill members of the gut microbiota ('gut commensals') and reduce competition for nutrients, making these nutrients available to support the growth of pathogens. This disturbance can lead to the growth and expansion of pathogens within the intestine (including antibiotic-resistant pathogens), where these pathogens can exploit the absence of competitors and the nutrient-enriched gut environment. In this review, we discuss nutrient competition between the gut microbiota and pathogens. We also provide an overview of how nutrient competition can be harnessed to support the design of next-generation microbiome therapeutics to restrict the growth of pathogens and prevent the development of invasive infections.
Collapse
Affiliation(s)
- Victoria Horrocks
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Olivia G. King
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Alexander Y. G. Yip
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Inês Melo Marques
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Julie A. K. McDonald
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
9
|
Arzamasov AA, Osterman AL. Milk glycan metabolism by intestinal bifidobacteria: insights from comparative genomics. Crit Rev Biochem Mol Biol 2022; 57:562-584. [PMID: 36866565 PMCID: PMC10192226 DOI: 10.1080/10409238.2023.2182272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Bifidobacteria are early colonizers of the human neonatal gut and provide multiple health benefits to the infant, including inhibiting the growth of enteropathogens and modulating the immune system. Certain Bifidobacterium species prevail in the gut of breastfed infants due to the ability of these microorganisms to selectively forage glycans present in human milk, specifically human milk oligosaccharides (HMOs) and N-linked glycans. Therefore, these carbohydrates serve as promising prebiotic dietary supplements to stimulate the growth of bifidobacteria in the guts of children suffering from impaired gut microbiota development. However, the rational formulation of milk glycan-based prebiotics requires a detailed understanding of how bifidobacteria metabolize these carbohydrates. Accumulating biochemical and genomic data suggest that HMO and N-glycan assimilation abilities vary remarkably within the Bifidobacterium genus, both at the species and strain levels. This review focuses on the delineation and genome-based comparative analysis of differences in respective biochemical pathways, transport systems, and associated transcriptional regulatory networks, providing a foundation for genomics-based projection of milk glycan utilization capabilities across a rapidly growing number of sequenced bifidobacterial genomes and metagenomic datasets. This analysis also highlights remaining knowledge gaps and suggests directions for future studies to optimize the formulation of milk-glycan-based prebiotics that target bifidobacteria.
Collapse
Affiliation(s)
- Aleksandr A Arzamasov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
10
|
Geng J, Sui Z, Dou W, Miao Y, Wang T, Wei X, Chen S, Zhang Z, Xiao J, Huang D. 16S rRNA Gene Sequencing Reveals Specific Gut Microbes Common to Medicinal Insects. Front Microbiol 2022; 13:892767. [PMID: 35651499 PMCID: PMC9149300 DOI: 10.3389/fmicb.2022.892767] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022] Open
Abstract
Insects have a long history of being used in medicine, with clear primary and secondary functions and less side effects, and the study and exploitation of medicinal insects have received increasing attention. Insects gut microbiota and their metabolites play an important role in protecting the hosts from other potentially harmful microbes, providing nutrients, promoting digestion and degradation, and regulating growth and metabolism of the hosts. However, there are still few studies linking the medicinal values of insects with their gut microbes. In this study, we focused on the specific gut microbiota common to medicinal insects, hoping to trace the potential connection between medicinal values and gut microbes of medicinal insects. Based on 16S rRNA gene sequencing data, we compared the gut microbiota of medicinal insects [Periplaneta americana, Protaetia (Liocola) brevitarsis (Lewis) and Musca domestica], in their medicinal stages, and non-medicinal insects (Hermetia illucens L., Tenebrio molitor, and Drosophila melanogaster), and found that the intestinal microbial richness of medicinal insects was higher, and there were significant differences in the microbial community structure between the two groups. We established a model using a random-forest method to preliminarily screen out several types of gut microbiota common to medicinal insects that may play medicinal values: Parabacteroides goldsteinii, Lactobacillus dextrinicus, Bifidobacterium longum subsp. infantis (B. infantis), and Vagococcus carniphilus. In particular, P. goldsteinii and B. infantis were most probably involved in the anti-inflammatory effects of medicinal insects. Our results revealed an association between medicinal insects and their gut microbes, providing new development directions and possibly potential tools for utilizing microbes to enhance the medicinal efficacy of medicinal insects.
Collapse
Affiliation(s)
- Jin Geng
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhuoxiao Sui
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihao Dou
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yunheng Miao
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Tao Wang
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xunfan Wei
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Sicong Chen
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zongqi Zhang
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jinhua Xiao
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dawei Huang
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
11
|
Button JE, Autran CA, Reens AL, Cosetta CM, Smriga S, Ericson M, Pierce JV, Cook DN, Lee ML, Sun AK, Alousi AM, Koh AY, Rechtman DJ, Jenq RR, McKenzie GJ. Dosing a synbiotic of human milk oligosaccharides and B. infantis leads to reversible engraftment in healthy adult microbiomes without antibiotics. Cell Host Microbe 2022; 30:712-725.e7. [PMID: 35504279 DOI: 10.1016/j.chom.2022.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 11/30/2022]
Abstract
Predictable and sustainable engraftment of live biotherapeutic products into the human gut microbiome is being explored as a promising way to modulate the human gut microbiome. We utilize a synbiotic approach pairing the infant gut microbe Bifidobacterium longum subspecies infantis (B. infantis) and human milk oligosaccharides (HMO). B. infantis, which is typically absent in adults, engrafts into healthy adult microbiomes in an HMO-dependent manner at a relative abundance of up to 25% of the bacterial population without antibiotic pretreatment or adverse effects. Corresponding changes in metabolites are detected. Germ-free mice transplanted with dysbiotic human microbiomes also successfully engraft with B. infantis in an HMO-dependent manner, and the synbiotic augments butyrate levels both in this in vivo model and in in vitro cocultures of the synbiotic with specific Firmicutes species. Finally, the synbiotic inhibits the growth of enteropathogens in vitro. Our findings point to a potential safe mechanism for ameliorating dysbioses characteristic of numerous human diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Adam K Sun
- Prolacta Bioscience, Duarte, CA 91010, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Robert R Jenq
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
12
|
Oladeinde A, Abdo Z, Zwirzitz B, Woyda R, Lakin SM, Press MO, Cox NA, Thomas JC, Looft T, Rothrock MJ, Zock G, Plumblee Lawrence J, Cudnik D, Ritz C, Aggrey SE, Liachko I, Grove JR, Wiersma C. Litter Commensal Bacteria Can Limit the Horizontal Gene Transfer of Antimicrobial Resistance to Salmonella in Chickens. Appl Environ Microbiol 2022; 88:e0251721. [PMID: 35416680 PMCID: PMC9107613 DOI: 10.1128/aem.02517-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/06/2022] [Indexed: 12/15/2022] Open
Abstract
Fostering a "balanced" gut microbiome through the administration of beneficial microbes that can competitively exclude pathogens has gained a lot of attention and use in human and animal medicine. However, little is known about how microbes affect the horizontal gene transfer of antimicrobial resistance (AMR). To shed more light on this question, we challenged neonatal broiler chicks raised on reused broiler chicken litter-a complex environment made up of decomposing pine shavings, feces, uric acid, feathers, and feed-with Salmonella enterica serovar Heidelberg (S. Heidelberg), a model pathogen. Neonatal chicks challenged with S. Heidelberg and raised on reused litter were more resistant to S. Heidelberg cecal colonization than chicks grown on fresh litter. Furthermore, chicks grown on reused litter were at a lower risk of colonization with S. Heidelberg strains that encoded AMR on IncI1 plasmids. We used 16S rRNA gene sequencing and shotgun metagenomics to show that the major difference between chicks grown on fresh litter and those grown on reused litter was the microbiome harbored in the litter and ceca. The microbiome of reused litter samples was more uniform and enriched in functional pathways related to the biosynthesis of organic and antimicrobial molecules than that in fresh litter samples. We found that Escherichia coli was the main reservoir of plasmids encoding AMR and that the IncI1 plasmid was maintained at a significantly lower copy per cell in reused litter compared to fresh litter. These findings support the notion that commensal bacteria play an integral role in the horizontal transfer of plasmids encoding AMR to pathogens like Salmonella. IMPORTANCE Antimicrobial resistance spread is a worldwide health challenge, stemming in large part from the ability of microorganisms to share their genetic material through horizontal gene transfer. To address this issue, many countries and international organizations have adopted a One Health approach to curtail the proliferation of antimicrobial-resistant bacteria. This includes the removal and reduction of antibiotics used in food animal production and the development of alternatives to antibiotics. However, there is still a significant knowledge gap in our understanding of how resistance spreads in the absence of antibiotic selection and the role commensal bacteria play in reducing antibiotic resistance transfer. In this study, we show that commensal bacteria play a key role in reducing the horizontal gene transfer of antibiotic resistance to Salmonella, provide the identity of the bacterial species that potentially perform this function in broiler chickens, and also postulate the mechanism involved.
Collapse
Affiliation(s)
| | - Zaid Abdo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Benjamin Zwirzitz
- Institute of Food Science, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Reed Woyda
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Steven M. Lakin
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | | | - Nelson A. Cox
- U.S. National Poultry Research Center, USDA—ARS, Athens, Georgia, USA
| | - Jesse C. Thomas
- Division of STD Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Torey Looft
- National Animal Disease Center, USDA—ARS, Ames, Iowa, USA
| | | | - Gregory Zock
- Department of Poultry Science, University of Georgia, Athens, Georgia, USA
| | | | - Denice Cudnik
- U.S. National Poultry Research Center, USDA—ARS, Athens, Georgia, USA
| | - Casey Ritz
- Department of Poultry Science, University of Georgia, Athens, Georgia, USA
| | - Samuel E. Aggrey
- Department of Poultry Science, University of Georgia, Athens, Georgia, USA
| | | | | | - Crystal Wiersma
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
13
|
Taft DH, Lewis ZT, Nguyen N, Ho S, Masarweh C, Dunne-Castagna V, Tancredi DJ, Huda MN, Stephensen CB, Hinde K, von Mutius E, Kirjavainen PV, Dalphin JC, Lauener R, Riedler J, Smilowitz JT, German JB, Morrow AL, Mills DA. Bifidobacterium Species Colonization in Infancy: A Global Cross-Sectional Comparison by Population History of Breastfeeding. Nutrients 2022; 14:nu14071423. [PMID: 35406036 PMCID: PMC9003546 DOI: 10.3390/nu14071423] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Bifidobacterium species are beneficial and dominant members of the breastfed infant gut microbiome; however, their health benefits are partially species-dependent. Here, we characterize the species and subspecies of Bifidobacterium in breastfed infants around the world to consider the potential impact of a historic dietary shift on the disappearance of B. longum subsp. infantis in some populations. Across populations, three distinct patterns of Bifidobacterium colonization emerged: (1) The dominance of Bifidobacterium longum subspecies infantis, (2) prevalent Bifidobacterium of multiple species, and (3) the frequent absence of any Bifidobacterium. These patterns appear related to a country’s history of breastfeeding, with infants in countries with historically high rates of long-duration breastfeeding more likely to be colonized by B. longum subspecies infantis compared with infants in countries with histories of shorter-duration breastfeeding. In addition, the timing of infant colonization with B. longum subsp. infantis is consistent with horizontal transmission of this subspecies, rather than the vertical transmission previously reported for other Bifidobacterium species. These findings highlight the need to consider historical and cultural influences on the prevalence of gut commensals and the need to understand epidemiological transmission patterns of Bifidobacterium and other major commensals.
Collapse
Affiliation(s)
- Diana H. Taft
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - Zachery T. Lewis
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Nhu Nguyen
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Steve Ho
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Chad Masarweh
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Vanessa Dunne-Castagna
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Daniel J. Tancredi
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - M. Nazmul Huda
- US Department of Agriculture, Western Human Nutrition Research Center, Davis, CA 95616, USA; (M.N.H.); (C.B.S.)
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Charles B. Stephensen
- US Department of Agriculture, Western Human Nutrition Research Center, Davis, CA 95616, USA; (M.N.H.); (C.B.S.)
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Katie Hinde
- Center for Evolution and Medicine, School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85281, USA;
| | - Erika von Mutius
- Dr. von Hauner Children’s Hospital, Ludwig Maximilian University, 80337 Munich, Germany;
- Institute for Asthma and Allergy Prevention, Helmholtz Centre Munich, 85764 Neuherberg, Germany
| | - Pirkka V. Kirjavainen
- Environment Health Unit, National Institute for Health and Welfare, 70210 Kuopio, Finland;
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jean-Charles Dalphin
- Department of Respiratory Disease, UMR/CNRS 6249 Chrono-Environment, University Hospital of Besançon, F-25000 Besançon, France;
| | - Roger Lauener
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland;
- Children’s Hospital of Eastern Switzerland, 9000 St. Gallen, Switzerland
| | - Josef Riedler
- Children’s Hospital Schwarzach, 5620 Schwarzach, Austria;
| | - Jennifer T. Smilowitz
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - J. Bruce German
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - Ardythe L. Morrow
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
- Correspondence: (A.L.M.); (D.A.M.); Tel.: +1-513-558-0809 (A.L.M.); +1-530-754-7821 (D.A.M.)
| | - David A. Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Department of Viticulture and Enology, University of California Davis, Davis, CA 95616, USA
- Correspondence: (A.L.M.); (D.A.M.); Tel.: +1-513-558-0809 (A.L.M.); +1-530-754-7821 (D.A.M.)
| |
Collapse
|
14
|
Lueschow SR, Boly TJ, Frese SA, Casaburi G, Mitchell RD, Henrick BM, McElroy SJ. Bifidobacterium longum Subspecies infantis Strain EVC001 Decreases Neonatal Murine Necrotizing Enterocolitis. Nutrients 2022; 14:495. [PMID: 35276854 PMCID: PMC8839161 DOI: 10.3390/nu14030495] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 01/04/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a disease mainly of preterm infants with a 30-50% mortality rate and long-term morbidities for survivors. Treatment strategies are limited and have not improved in decades, prompting research into prevention strategies, particularly with probiotics. Recent work with the probiotic B. infantis EVC001 suggests that this organism may generate a more appropriate microbiome for preterm infants who generally have inappropriate gut colonization and inflammation, both risk factors for NEC. Experimental NEC involving Paneth cell disruption in combination with bacterial dysbiosis or formula feeding was induced in P14-16 C57Bl/6 mice with or without gavaged B. infantis. Following completion of the model, serum, small intestinal tissue, the cecum, and colon were harvested to examine inflammatory cytokines, injury, and the microbiome, respectively. EVC001 treatment significantly decreased NEC in a bacterial dysbiosis dependent model, but this decrease was model-dependent. In the NEC model dependent on formula feeding, no difference in injury was observed, but trending to significant differences was observed in serum cytokines. EVC001 also improved wound closure at six and twelve hours compared to the sham control in intestinal epithelial monolayers. These findings suggest that B. infantis EVC001 can prevent experimental NEC through anti-inflammatory and epithelial barrier restoration properties.
Collapse
Affiliation(s)
- Shiloh R. Lueschow
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Timothy J. Boly
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven A. Frese
- Department of Nutrition, University of Nevada, Reno, NV 89557, USA;
| | - Giorgio Casaburi
- Department of Bioinformatics, Metabiomics, Carlsbad, CA 92008, USA;
| | - Ryan D. Mitchell
- Evolve Biosystems, Inc., Davis, CA 95618, USA; (R.D.M.); (B.M.H.)
| | - Bethany M. Henrick
- Evolve Biosystems, Inc., Davis, CA 95618, USA; (R.D.M.); (B.M.H.)
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - Steven J. McElroy
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
15
|
Casaburi G, Wei J, Kazi S, Liu J, Wang K, Tao GZ, Lin PY, Dunn JCY, Henrick BM, Frese SA, Sylvester KG. Metabolic model of necrotizing enterocolitis in the premature newborn gut resulting from enteric dysbiosis. Front Pediatr 2022; 10:893059. [PMID: 36081629 PMCID: PMC9445129 DOI: 10.3389/fped.2022.893059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of premature newborn morbidity and mortality. The clinical features of NEC consistently include prematurity, gut dysbiosis and enteral inflammation, yet the pathogenesis remains obscure. Herein we combine metagenomics and targeted metabolomics, with functional in vivo and in vitro assessment, to define a novel molecular mechanism of NEC. One thousand six hundred and forty seven publicly available metagenomics datasets were analyzed (NEC = 245; healthy = 1,402) using artificial intelligence methodologies. Targeted metabolomic profiling was used to quantify the concentration of specified fecal metabolites at NEC onset (n = 8), during recovery (n = 6), and in age matched controls (n = 10). Toxicity assays of discovered metabolites were performed in vivo in mice and in vitro using human intestinal epithelial cells. Metagenomic and targeted metabolomic analyses revealed significant differences in pyruvate fermentation pathways and associated intermediates. Notably, the short chain fatty acid formate was elevated in the stool of NEC patients at disease onset (P = 0.005) dissipated during recovery (P = 0.02) and positively correlated with degree of intestinal injury (r 2 = 0.86). In vitro, formate caused enterocyte cytotoxicity in human cells through necroptosis (P < 0.01). In vivo, luminal formate caused significant dose and development dependent NEC-like injury in newborn mice. Enterobacter cloacae and Klebsiella pneumoniae were the most discriminatory taxa related to NEC dysbiosis and increased formate production. Together, these data suggest a novel biochemical mechanism of NEC through the microbial production of formate. Clinical efforts to prevent NEC should focus on reducing the functional consequences of newborn gut dysbiosis associated metabolic pathways.
Collapse
Affiliation(s)
| | - Jingjing Wei
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of Pediatrics, Shanxi Medical University, Taiyuan, China
| | - Sufyan Kazi
- Evolve Biosystems, Inc., Davis, CA, United States
| | - Junlin Liu
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of General Surgery, The People's Hospital of Liuyang City, Liuyang, China
| | - Kewei Wang
- Department of Surgery, Stanford University, Stanford, CA, United States.,Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Guo-Zhong Tao
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Po-Yu Lin
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - James C Y Dunn
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Bethany M Henrick
- Evolve Biosystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Steven A Frese
- Evolve Biosystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States.,Department of Nutrition, University of Nevada Reno, Reno, NV, United States
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
16
|
Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, Olin A, Wang J, Mikes J, Tan Z, Chen Y, Ehrlich AM, Bernhardsson AK, Mugabo CH, Ambrosiani Y, Gustafsson A, Chew S, Brown HK, Prambs J, Bohlin K, Mitchell RD, Underwood MA, Smilowitz JT, German JB, Frese SA, Brodin P. Bifidobacteria-mediated immune system imprinting early in life. Cell 2021; 184:3884-3898.e11. [PMID: 34143954 DOI: 10.1016/j.cell.2021.05.030] [Citation(s) in RCA: 402] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/19/2021] [Accepted: 05/19/2021] [Indexed: 02/08/2023]
Abstract
Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon β (IFNβ) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.
Collapse
Affiliation(s)
- Bethany M Henrick
- Evolve BioSystems, Inc., Davis, CA 95618, USA; Department of Food Science and Technology, University of Nebraska, Lincoln, Lincoln, NE 68588-6205, USA.
| | - Lucie Rodriguez
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Christian Pou
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ewa Henckel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Aron Arzoomand
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Axel Olin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Jun Wang
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Jaromir Mikes
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ziyang Tan
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Yang Chen
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | | | - Anna Karin Bernhardsson
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Constantin Habimana Mugabo
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ylva Ambrosiani
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Anna Gustafsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | | | | | | | - Kajsa Bohlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | | | - Mark A Underwood
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Pediatrics, University of California Davis Children's Hospital, Sacramento, CA 95817, USA
| | - Jennifer T Smilowitz
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, Davis, CA 95616, USA
| | - J Bruce German
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, Davis, CA 95616, USA
| | - Steven A Frese
- Department of Food Science and Technology, University of Nebraska, Lincoln, Lincoln, NE 68588-6205, USA; Department of Nutrition, University of Nevada, Reno, Reno, NV 89557, USA
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden; Pediatric Rheumatology, Karolinska University Hospital, 17176 Solna, Sweden.
| |
Collapse
|
17
|
Dimitratos SM, Brown H, Shafizadeh T, Kazi S, Altmann T, Ostrer B. Symptomatic relief from at-home use of activated Bifidobacterium infantis EVC001 probiotic in infants: results from a consumer survey on the effects on diaper rash, colic symptoms, and sleep. Benef Microbes 2021; 12:27-34. [PMID: 34057053 DOI: 10.3920/bm2020.0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The gut microbiome during infancy is directly involved in the digestion of human milk, development of the immune system, and long-term health outcomes. Gut dysbiosis in early life has been linked to multiple short-term ailments, from diaper dermatitis and poor stooling habits, to poor sleep and fussiness, with mixed results in the scientific literature on the efficacy of probiotics for symptom resolution. Despite the growing interest in probiotics for consumer use, observed symptomatic relief is rarely documented. This study aims to evaluate observed symptomatic relief from at-home use of activated Bifidobacterium infantis EVC001 in infants. Consumer feedback was collected over a 2-year period via a 30-day post-purchase online survey of B. infantis EVC001 (Evivo®) customers. Outcome measures included observed changes in diaper rash, symptoms of colic, and sleep behaviours in infants fed B. infantis EVC001. A total of 1,621 respondents completed the survey. Before purchasing B. infantis EVC001, the majority of respondents visited the product website, researched infant probiotics online, or consulted with their doctor or other healthcare professional. Of the participants whose infants had ever experienced diaper rash, 72% (n=448) reported improvements, and 57% of those reported complete resolution of this problem. Of those who responded to questions about gassiness/fussiness, naptime sleep, and night-time sleep behaviours, 63% (n=984), 33% (n=520), and 52% (n=806) reported resolution or improvements, respectively. Although clinical data regarding probiotic use are often inconclusive for symptom resolution, home use of B. infantis EVC001 in infants improved diaper rash, gassiness/fussiness, and sleep quality within the first week of use in a significant number of respondents who engaged in a voluntary post-purchase survey. These outcomes may be a result of the unique genetic capacity of B. infantis EVC001 to colonise the infant gut highlighting the importance of strain selection in evaluating the effects of probiotic products.
Collapse
Affiliation(s)
- S M Dimitratos
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - H Brown
- Evolve BioSystems, Inc, Davis, CA 95618, USA
| | | | - S Kazi
- Evolve BioSystems, Inc, Davis, CA 95618, USA
| | - T Altmann
- Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.,Calabasas Pediatrics Wellness Center, Calabasas, CA 91302, USA
| | - B Ostrer
- Evolve BioSystems, Inc, Davis, CA 95618, USA
| |
Collapse
|
18
|
Duman H, Kaplan M, Arslan A, Sahutoglu AS, Kayili HM, Frese SA, Karav S. Potential Applications of Endo-β- N-Acetylglucosaminidases From Bifidobacterium longum Subspecies infantis in Designing Value-Added, Next-Generation Infant Formulas. Front Nutr 2021; 8:646275. [PMID: 33898500 PMCID: PMC8063050 DOI: 10.3389/fnut.2021.646275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Human milk is the optimal source of infant nutrition. Among many other health benefits, human milk can stimulate the development of a Bifidobacterium-rich microbiome through human milk oligosaccharides (HMOs). In recent years, the development of novel formulas has placed particular focus on incorporating some of the beneficial functional properties of human milk. These include adding specific glycans aimed to selectively stimulate the growth of Bifidobacterium. However, the bifidogenicity of human milk remains unparalleled. Dietary N-glycans are carbohydrate structures conjugated to a wide variety of glycoproteins. These glycans have a remarkable structural similarity to HMOs and, when released, show a strong bifidogenic effect. This review discusses the biocatalytic potential of the endo-β-N-acetylglucosaminidase enzyme (EndoBI-1) from Bifidobacterium longum subspecies infantis (B. infantis), in releasing N-glycans inherently present in infant formula as means to increase the bifidogenicity of infant formula. Finally, the potential implications for protein deglycosylation with EndoBI-1 in the development of value added, next-generation formulas are discussed from a technical perspective.
Collapse
Affiliation(s)
- Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Merve Kaplan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Ayşenur Arslan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | | | - Haci Mehmet Kayili
- Department of Biomedical Engineering, Karabuk University, Karabük, Turkey
| | - Steven A Frese
- Department of Nutrition, University of Nevada, Reno, NV, United States.,Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
19
|
Duar RM, Kyle D, Tribe RM. Reintroducing B. infantis to the cesarean-born neonate: an ecologically sound alternative to "vaginal seeding". FEMS Microbiol Lett 2021; 367:5739918. [PMID: 32068827 PMCID: PMC7182402 DOI: 10.1093/femsle/fnaa032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
There is a burgeoning literature highlighting differences in health outcomes between babies born vaginally and by caesarean section (c-section) This has led to the suggestion that infants born by c-section may benefit from vaginal swabbing/seeding. Here, we discuss from an ecological perspective that it is gut-adapted, not vagina-adapted microbes that are likely to take up residence in the gut and have the most beneficial impact on the developing neonate. Further, we caution the practice of 'vaginal seeding' may be potentially unsafe and also give parents and health professionals a false sense of action in restoring the infant gut microbiome following c-section. Instead, we argue that restoring B. longum subsp. infantis, which has evolved to colonize the infant gut, is a safe and ecologically-sound approach to restoring the gut microbiome of infants born by c-section.
Collapse
Affiliation(s)
| | - David Kyle
- Evolve BioSytems, Inc. Davis, California, 95618 USA
| | - Rachel M Tribe
- Dept. of Women and Children's Health, School of Life Course Sciences, St Thomas' Hospital, London SE1 7EH UK
| |
Collapse
|
20
|
Nguyen M, Holdbrooks H, Mishra P, Abrantes MA, Eskew S, Garma M, Oca CG, McGuckin C, Hein CB, Mitchell RD, Kazi S, Chew S, Casaburi G, Brown HK, Frese SA, Henrick BM. Impact of Probiotic B. infantis EVC001 Feeding in Premature Infants on the Gut Microbiome, Nosocomially Acquired Antibiotic Resistance, and Enteric Inflammation. Front Pediatr 2021; 9:618009. [PMID: 33665175 PMCID: PMC7921802 DOI: 10.3389/fped.2021.618009] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Preterm birth is a major determinant of neonatal survival and morbidity, but the gut microbiome and associated enteric inflammation are also key factors in neonatal development and the risk of associated morbidities. We prospectively and longitudinally followed two cohorts of preterm infants, one of which was fed activated Bifidobacterium longum subsp. infantis (B. infantis) EVC001 8 × 109 CFU daily, and the other was not fed a probiotic. Hospital feeding protocol assigned all infants born at <1500 g and/or < 32 weeks corrected gestational age to the probiotic feeding protocol, whereas infants born at >1500 g and/or >32 weeks corrected gestational age were not fed a probiotic. Fecal samples were opportunistically collected from 77 infants throughout the hospital stay, and subjected to shotgun metagenomic sequencing and quantification of enteric inflammation. De-identified metadata was collected from patient medical records. Results: The gut microbiome of preterm infants was typified by a high abundance of Enterobacteriaceae and/or Staphylococcaceae, and multivariate modeling identified the probiotic intervention, rather than degree of prematurity, day of life, or other clinical interventions, as the primary source of change in the gut microbiome. Among infants fed B. infantis EVC001, a high abundance of total Bifidobacteriaceae developed rapidly, the majority of which was B. infantis confirmed via subspecies-specific qPCR. Associated with this higher abundance of Bifidobacteriaceae, we found increased functional capacity for utilization of human milk oligosaccharides (HMOs), as well as reduced abundance of antibiotic resistance genes (ARGs) and the taxa that harbored them. Importantly, we found that infants fed B. infantis EVC001 exhibited diminished enteric inflammation, even when other clinical variables were accounted for using multivariate modeling. Conclusion: These results provide an important observational background for probiotic use in a NICU setting, and describe the clinical, physiological, and microbiome-associated improvements in preterm infants associated with B. infantis EVC001 feeding.
Collapse
Affiliation(s)
- Marielle Nguyen
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Heaven Holdbrooks
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Prasanthi Mishra
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Maria A. Abrantes
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Sherri Eskew
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Mariajamiela Garma
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | - Cyr-Geraurd Oca
- Neonatology, Kaiser Permanente Orange County, Anaheim, CA, United States
| | | | | | | | - Sufyan Kazi
- Evolve Biosystems Inc., Davis, CA, United States
| | | | | | | | - Steven A. Frese
- Evolve Biosystems Inc., Davis, CA, United States
- Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
- Department of Nutrition, University of Nevada, Reno, NV, United States
| | - Bethany M. Henrick
- Evolve Biosystems Inc., Davis, CA, United States
- Department of Food Science and Technology, University of Nebraska Lincoln, Lincoln, NE, United States
| |
Collapse
|
21
|
Casaburi G, Duar RM, Brown H, Mitchell RD, Kazi S, Chew S, Cagney O, Flannery RL, Sylvester KG, Frese SA, Henrick BM, Freeman SL. Metagenomic insights of the infant microbiome community structure and function across multiple sites in the United States. Sci Rep 2021; 11:1472. [PMID: 33479326 PMCID: PMC7820601 DOI: 10.1038/s41598-020-80583-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome plays an important role in early life, protecting newborns from enteric pathogens, promoting immune system development and providing key functions to the infant host. Currently, there are limited data to broadly assess the status of the US healthy infant gut microbiome. To address this gap, we performed a multi-state metagenomic survey and found high levels of bacteria associated with enteric inflammation (e.g. Escherichia, Klebsiella), antibiotic resistance genes, and signatures of dysbiosis, independent of location, age, and diet. Bifidobacterium were less abundant than generally expected and the species identified, including B. breve, B. longum and B. bifidum, had limited genetic capacity to metabolize human milk oligosaccharides (HMOs), while B. infantis strains with a complete capacity for HMOs utilization were found to be exceptionally rare. Considering microbiome composition and functional capacity, this survey revealed a previously unappreciated dysbiosis that is widespread in the contemporary US infant gut microbiome.
Collapse
Affiliation(s)
| | | | | | | | - Sufyan Kazi
- Evolve BioSystems, Inc., Davis, CA, 95618, USA
| | | | - Orla Cagney
- Evolve BioSystems, Inc., Davis, CA, 95618, USA
| | | | | | - Steven A Frese
- Evolve BioSystems, Inc., Davis, CA, 95618, USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, 68588, USA
- Department of Nutrition, University of Nevada, Reno, Reno, NV, 89557, USA
| | - Bethany M Henrick
- Evolve BioSystems, Inc., Davis, CA, 95618, USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, 68588, USA
| | | |
Collapse
|
22
|
Duar RM, Casaburi G, Mitchell RD, Scofield LN, Ortega Ramirez CA, Barile D, Henrick BM, Frese SA. Comparative Genome Analysis of Bifidobacterium longum subsp. infantis Strains Reveals Variation in Human Milk Oligosaccharide Utilization Genes among Commercial Probiotics. Nutrients 2020; 12:nu12113247. [PMID: 33114073 PMCID: PMC7690671 DOI: 10.3390/nu12113247] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/23/2022] Open
Abstract
Dysbiosis is associated with acute and long-term consequences for neonates. Probiotics can be effective in limiting the growth of bacteria associated with dysbiosis and promoting the healthy development of the infant microbiome. Given its adaptation to the infant gut, and promising data from animal and in vitro models, Bifidobacterium longum subsp. infantis is an attractive candidate for use in infant probiotics. However, strain-level differences in the ability of commercialized strains to utilize human milk oligosaccharides (HMOs) may have implications in the performance of strains in the infant gut. In this study, we characterized twelve B. infantis probiotic strains and identified two main variants in one of the HMO utilization gene clusters. Some strains possessed the full repertoire of HMO utilization genes (H5-positive strains), while H5-negative strains lack an ABC-type transporter known to bind core HMO structures. H5-positive strains achieved significantly superior growth on lacto-N-tetraose and lacto-N-neotetraose. In vitro, H5-positive strains had a significant fitness advantage over H5-negative strains, which was also observed in vivo in breastfed infants. This work provides evidence of the functional implications of genetic differences among B. infantis strains and highlights that genotype and HMO utilization phenotype should be considered when selecting a strain for probiotic use in infants.
Collapse
Affiliation(s)
- Rebbeca M. Duar
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
| | - Giorgio Casaburi
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
| | - Ryan D. Mitchell
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
| | - Lindsey N.C. Scofield
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
| | - Camila A. Ortega Ramirez
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
| | - Daniela Barile
- Foods for Health Institute, University of California at Davis, Davis, CA 95616, USA;
- Department of Food Science and Technology, University of California at Davis, Davis, CA 95616, USA
| | - Bethany M. Henrick
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - Steven A. Frese
- Evolve BioSystems, Inc., Davis, CA 95618, USA; (R.M.D.); (G.C.); (R.D.M.); (L.N.C.S.); (C.A.O.R.); (B.M.H.)
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
- Correspondence: ; Tel.: +1-530-747-2045
| |
Collapse
|
23
|
Czajkowski MD, Vance DP, Frese SA, Casaburi G. GenCoF: a graphical user interface to rapidly remove human genome contaminants from metagenomic datasets. Bioinformatics 2020; 35:2318-2319. [PMID: 30475995 PMCID: PMC6596892 DOI: 10.1093/bioinformatics/bty963] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/07/2018] [Accepted: 11/23/2018] [Indexed: 11/22/2022] Open
Abstract
Summary The removal of human genomic reads from shotgun metagenomic sequencing is a critical step in protecting subject privacy. Freely available tools addressing this issue require advanced programing knowledge or are limited by analytical time and data load due to their server-based nature. Here, we compared the most cited tools for host-DNA removal using synthetic and real metagenomic datasets. Then, we integrated the most efficient pipeline in a graphical user interface to make these tools available without command line use. This interface, GenCoF, rapidly removes human genome contaminants from metagenomic datasets. Additionally, the tool offers quality-filtering, data reduction and interactive modification of any parameter in order to customize the analysis. GenCoF offers both quality and host-associated filtering in a non-commercial, freely available tool in a local, interactive and easy-to-use interface. Availability and implementation GenCoF is freely available (under a GPL license) for Mac OS and Linux at https://github.com/MattCzajkowski/GenCoF. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | | | - Steven A Frese
- Evolve BioSystems, Inc., Davis, CA, USA.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
| | | |
Collapse
|
24
|
Ford EL, Underwood MA, German JB. Helping Mom Help Baby: Nutrition-Based Support for the Mother-Infant Dyad During Lactation. Front Nutr 2020; 7:54. [PMID: 32373623 PMCID: PMC7186439 DOI: 10.3389/fnut.2020.00054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Lactation and breastfeeding support the short- and long-term health of both mother and infant, yet the success of these processes depend upon individual and combined factors of the pair. Complications during pregnancy and delivery greatly affect the likelihood that a mother will be capable of breastfeeding for at least the recommended 6 months. Guidelines for women regarding postpartum diet and lifestyle management also fail to reflect the diversity of mother-infant pairs and their circumstances. In our analysis of the literature, we have identified a categorical deficit in modern scientific discourse regarding human lactation; namely, that postpartum involves full-body contribution of resources and thus requires the application of nutrition from a systemic perspective.
Collapse
Affiliation(s)
- Erin L Ford
- Department of Food Science and Technology, Foods for Health Institute, University of California, Davis, Davis, CA, United States
| | | | - J Bruce German
- Department of Food Science and Technology, Foods for Health Institute, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
25
|
Duar RM, Henrick BM, Casaburi G, Frese SA. Integrating the Ecosystem Services Framework to Define Dysbiosis of the Breastfed Infant Gut: The Role of B. infantis and Human Milk Oligosaccharides. Front Nutr 2020; 7:33. [PMID: 32346537 PMCID: PMC7171047 DOI: 10.3389/fnut.2020.00033] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Abstract
Mounting evidence supports a connection between the composition of the infant gut microbiome and long-term health. In fact, aberrant microbiome compositions during key developmental windows in early life are associated with increased disease risk; therefore, making pertinent modifications to the microbiome during infancy offers significant promise to improve human health. There is growing support for integrating the concept of ecosystem services (the provision of benefits from ecosystems to humans) in linking specific microbiome functions to human well-being. This framework is widely applied in conservation efforts of macro-ecosystems and offers a systematic approach to guide restoration actions aimed to recover critical ecological functions. The aim of this work is to apply the ecosystem services framework to integrate recent studies demonstrating stable alteration of the gut microbiome of breastfed infants when Bifidobacterium longum subsp. infantis EVC001, a gut symbiont capable of efficiently utilizing human milk oligosaccharides into organic acids that are beneficial for the infant and lower intestinal pH, is reintroduced. Additionally, using examples from the literature we illustrate how the absence of B. infantis results in diminished ecosystem services, which may be associated with health consequences related to immune and metabolic disorders. Finally, we propose a model by which infant gut dysbiosis can be defined as a reduction in ecosystem services supplied to the host by the gut microbiome rather than merely changes in diversity or taxonomic composition. Given the increased interest in targeted microbiome modification therapies to decrease acute and chronic disease risk, the model presented here provides a framework to assess the effectiveness of such strategies from a host-centered perspective.
Collapse
Affiliation(s)
| | - Bethany M Henrick
- Evolve BioSystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | | | - Steven A Frese
- Evolve BioSystems, Inc., Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| |
Collapse
|
26
|
Colonization Resistance in the Infant Gut: The Role of B. infantis in Reducing pH and Preventing Pathogen Growth. High Throughput 2020; 9:ht9020007. [PMID: 32230716 PMCID: PMC7349288 DOI: 10.3390/ht9020007] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/17/2022] Open
Abstract
Over the past century, there has been a steady increase in the stool pH of infants from industrialized countries. Analysis of historical data revealed a strong association between abundance of Bifidobacterium in the gut microbiome of breasted infants and stool pH, suggesting that this taxon plays a key role in determining the pH in the gut. Bifidobacterium longum subsp. infantis is uniquely equipped to metabolize human milk oligosaccharides (HMO) from breastmilk into acidic end products, mainly lactate and acetate. The presence of these acidic compounds in the infant gut is linked to a lower stool pH. Conversely, infants lacking B. infantis have a significantly higher stool pH, carry a higher abundance of potential pathogens and mucus-eroding bacteria in their gut microbiomes, and have signs of chronic enteric inflammation. This suggests the presence of B. infantis and low intestinal pH may be critical to maintaining a protective environment in the infant gut. Here, we summarize recent studies demonstrating that feeding B. infantis EVC001 to breastfed infants results in significantly lower fecal pH compared to controls and propose that low pH is one critical factor in preventing the invasion and overgrowth of harmful bacteria in the infant gut, a process known as colonization resistance.
Collapse
|
27
|
Casaburi G, Duar RM, Vance DP, Mitchell R, Contreras L, Frese SA, Smilowitz JT, Underwood MA. Early-life gut microbiome modulation reduces the abundance of antibiotic-resistant bacteria. Antimicrob Resist Infect Control 2019; 8:131. [PMID: 31423298 PMCID: PMC6693174 DOI: 10.1186/s13756-019-0583-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/22/2019] [Indexed: 01/21/2023] Open
Abstract
Background Antibiotic-resistant (AR) bacteria are a global threat. AR bacteria can be acquired in early life and have long-term sequelae. Limiting the spread of antibiotic resistance without triggering the development of additional resistance mechanisms is of immense clinical value. Here, we show how the infant gut microbiome can be modified, resulting in a significant reduction of AR genes (ARGs) and the potentially pathogenic bacteria that harbor them. Methods The gut microbiome was characterized using shotgun metagenomics of fecal samples from two groups of healthy, term breastfed infants. One group was fed B. infantis EVC001 in addition to receiving lactation support (n = 29, EVC001-fed), while the other received lactation support alone (n = 31, controls). Coliforms were isolated from fecal samples and genome sequenced, as well as tested for minimal inhibitory concentrations against clinically relevant antibiotics. Results Infants fed B. infantis EVC001 exhibited a change to the gut microbiome, resulting in a 90% lower level of ARGs compared to controls. ARGs that differed significantly between groups were predicted to confer resistance to beta lactams, fluoroquinolones, or multiple drug classes, the majority of which belonged to Escherichia, Clostridium, and Staphylococcus. Minimal inhibitory concentration assays confirmed the resistance phenotypes among isolates with these genes. Notably, we found extended-spectrum beta lactamases among healthy, vaginally delivered breastfed infants who had never been exposed to antibiotics. Conclusions Colonization of the gut of breastfed infants by a single strain of B. longum subsp. infantis had a profound impact on the fecal metagenome, including a reduction in ARGs. This highlights the importance of developing novel approaches to limit the spread of these genes among clinically relevant bacteria. Future studies are needed to determine whether colonization with B. infantis EVC001 decreases the incidence of AR infections in breastfed infants. Trial registration This clinical trial was registered at ClinicalTrials.gov, NCT02457338.
Collapse
Affiliation(s)
| | | | | | | | | | - Steven A. Frese
- Evolve Biosystems, Inc, Davis, CA 95618 USA
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588 USA
| | - Jennifer T. Smilowitz
- Department of Food Science and Technology, University of California, Davis, CA 95616 USA
- Foods for Health Institute, University of California, Davis, CA 95616 USA
| | - Mark A. Underwood
- Foods for Health Institute, University of California, Davis, CA 95616 USA
- Department of Pediatrics, UC Davis Children’s Hospital, Sacramento, CA 95817 USA
| |
Collapse
|
28
|
|
29
|
Abstract
PURPOSE OF REVIEW Probiotic administration to premature infants for the purpose of prevention of necrotizing enterocolitis is common in many parts of the world but uncommon in the United States. The present review will emphasize recent findings in support of routine administration of probiotics to this highly vulnerable population. RECENT FINDINGS Additional evidence from animal models describing mechanisms of protection of probiotics in the immature gut and updated meta-analyses of randomized placebo-controlled trials and observational cohorts are presented (now including more than 40 000 premature infants from countries across the globe). SUMMARY The preponderance of evidence suggests that probiotic administration to premature infants is well tolerated and decreases the risk of death, necrotizing enterocolitis, and sepsis. Further comparisons of probiotic administration to placebo are not likely to alter these conclusions. Rather, future work should focus on assurance of high-quality products with demonstrated purity and viability of probiotic microbes, and future clinical trials should focus on comparisons between high-quality products and doses.
Collapse
|
30
|
Identification of Differentiating Metabolic Pathways between Infant Gut Microbiome Populations Reveals Depletion of Function-Level Adaptation to Human Milk in the Finnish Population. mSphere 2019; 4:4/2/e00152-19. [PMID: 30894435 PMCID: PMC6429046 DOI: 10.1128/mspheredirect.00152-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Knowing the limitations of taxonomy-based research, there is an emerging need for the development of higher-resolution techniques. The significance of this research is demonstrated by the novel method used for the analysis of function-level metagenomes. BiomeScout—the presented technology—utilizes proprietary algorithms for the detection of differences between functionalities present in metagenomic samples. A variety of autoimmune and allergy events are becoming increasingly common, especially in Western countries. Some pieces of research link such conditions with the composition of microbiota during infancy. In this period, the predominant form of nutrition for gut microbiota is oligosaccharides from human milk (HMO). A number of gut-colonizing strains, such as Bifidobacterium and Bacteroides, are able to utilize HMO, but only some Bifidobacterium strains have evolved to digest the specific composition of human oligosaccharides. Differences in the proportions of the two genera that are able to utilize HMO have already been associated with the frequency of allergies and autoimmune diseases in the Finnish and the Russian populations. Our results show that differences in terms of the taxonomic annotation do not explain the reason for the differences in the Bifidobacterium/Bacteroides ratio between the Finnish and the Russian populations. In this paper, we present the results of function-level analysis. Unlike the typical workflow for gene abundance analysis, BiomeScout technology explains the differences in the Bifidobacterium/Bacteroides ratio. Our research shows the differences in the abundances of the two enzymes that are crucial for the utilization of short type 1 oligosaccharides. IMPORTANCE Knowing the limitations of taxonomy-based research, there is an emerging need for the development of higher-resolution techniques. The significance of this research is demonstrated by the novel method used for the analysis of function-level metagenomes. BiomeScout—the presented technology—utilizes proprietary algorithms for the detection of differences between functionalities present in metagenomic samples.
Collapse
|
31
|
Karav S, Casaburi G, Frese SA. Reduced colonic mucin degradation in breastfed infants colonized by Bifidobacterium longum subsp . infantis EVC001. FEBS Open Bio 2018; 8:1649-1657. [PMID: 30338216 PMCID: PMC6168692 DOI: 10.1002/2211-5463.12516] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
Mucin glycoproteins play an important role in protecting the gut epithelium by keeping gut microbes from direct contact with the gut epithelium while allowing for diffusion of small molecules from the lumen to the epithelium. The mucin glycocalyx can be degraded by gut bacteria such as Bacteroides and Akkermansia, but other bacteria, such as Bifidobacterium longum subsp. Infantis, cannot consume mucin glycans. Untargeted mass spectrometry profiles were compared to microbiome profiles to assess how different gut microbiomes affect colonic mucin degradation. Samples obtained from nine infants colonized by Bifidobacterium infantis EVC001 and from 10 infants colonized by higher levels of mucolytic taxa (controls), including Bacteroides, were compared. Previously performed untargeted nano‐high‐performance liquid chromatography‐chip/time‐of‐flight mass spectrometry was used to detect and quantify glycans originating from colonic mucin. Colonic mucin‐derived O‐glycans from control infants composed 37.68% (± 3.14% SD) of the total glycan structure pool, whereas colonic mucin‐derived O‐glycans made up of only 1.78% (± 0.038% SD) of the total in B. infantis EVC001 samples. The relative abundance of these colonic mucin‐derived O‐glycans in the total glycan pool was higher among control, 26.98% (± 8.48% SD), relative to B. infantis‐colonized infants, 1.68% (± 1.12% SD). Key taxa, such as Bacteroidaceae, were significantly and positively correlated with the abundance of these structures, while Bifidobacteriaceae were significantly and negatively associated with these structures. These results suggest that colonization of infants by B. infantis may diminish colonic glycan degradation and help maintain barrier function in the gastrointestinal tract of infants.
Collapse
Affiliation(s)
- Sercan Karav
- Department of Molecular Biology and Genetics Çanakkale Onsekiz Mart University Turkey
| | | | - Steven A Frese
- Evolve Biosystems, Inc. Davis CA USA.,Department of Food Science and Technology University of Nebraska Lincoln NE USA
| |
Collapse
|