151
|
Shen W, Qiu W, Liu Y, Liao W, Ma Y, He Y, Wang Z, Zhou H. Postnatal age is strongly correlated with the early development of the gut microbiome in preterm infants. Transl Pediatr 2021; 10:2313-2324. [PMID: 34733672 PMCID: PMC8506066 DOI: 10.21037/tp-21-367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The gut microbiome plays a potential role in clinical events in preterm infants and may affect their lateral development. Understanding the initial colonization of microbes in the gut, their early dynamic changes, and the major factors correlated with these changes would provide crucial information about the developmental process in early life. METHODS The present study enrolled 151 preterm infants and examined the longitudinal dynamics of their fecal microbiome profiles during the period of hospitalization using 16S ribosomal RNA gene sequencing. Random forest modeling was used to predict postnatal age (Age), postmenstrual age (PMA), and gestational age (GA), using gut microbiome features. RESULTS Principal coordinate analysis revealed that the gut microbiome of the preterm infants displayed an obvious time-dependent change pattern, which showed the strongest association with Age, followed by PMA, and a much weaker association with (GA). Random forest modeling further evidenced the time-dependent change pattern, with the Pearson's correlation coefficients between the actual values and the gut microbiome-predicted values being 0.68, 0.53, and 0.38 for postnatal, postmenstrual, and gestational age, respectively. The microbiome dynamism could be further divided into four Age stages, each with its own characteristic microbial taxa. The first 1-4 days (T1 stage) represented the meconium microbiome, with colonization of a high diversity of microbes before or during delivery. During 5-15 days (T2 stage), the gut microbiome of the preterm infants underwent a rapid turnover, in which microbial diversity declined, and stabilized afterward. Enterobacteriaceae, Enterococcaceae, Streptococcaceae, Staphylococcaceae, and Clostridiaceae were the major classes in the gut microbiome in the lateral stages of development (T3-T4 stage). CONCLUSIONS Postnatal age, rather than the gestational age, is significantly correlated with the gut microbiome of preterm infants, suggesting that clinical interventions contribute more to the early dynamics of gut microbiome in preterm infants than the natural development of the gut.
Collapse
Affiliation(s)
- Wei Shen
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Liu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weihua Liao
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Ma
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
152
|
Chen J, Li H, Hird SM, Chen MH, Xu W, Maas K, Cong X. Sex Differences in Gut Microbial Development of Preterm Infant Twins in Early Life: A Longitudinal Analysis. Front Cell Infect Microbiol 2021; 11:671074. [PMID: 34458157 PMCID: PMC8387566 DOI: 10.3389/fcimb.2021.671074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Infant gut microbiota plays a vital role in immune response, mediates neurobehavioral development and health maintenance. Studies of twins’ gut microbiota found that gut microbiota composition and diversity tend to be mature and stable with increasing postnatal age (PNA). Preterm infant gut microbiome shifts dramatically when they were staying in the neonatal intensive care unit (NICU). Compositions and shifting characteristics of gut microbiota among neonatal preterm twins and triplets during their early life are still unknown, which impedes a better understanding of the mechanism underpinning neurobehavioral development and precise intervention/health of preterm neonates. This longitudinal cohort study used a twins/triplets design to investigate the interaction of genetic (e.g., male vs. female) and environmental factors influencing the development of the gut microbiome in early life. We included 39 preterm infants, 12 were Female twins/triplets (Female T/T) including 3 twins pairs and 2 triplets, 12 were male twins (Male T) including 6 twins pairs, and 15 were mixed-sex twins/triplets (Mix T/T) including 6 twins pairs and 1 triplet (8 females and 7 males) during the first four weeks of NICU stay. Weekly gut microbiota patterns between females and males were compared by linear discriminant analysis (LDA) effect size (LEfSe). Metagenomics function of gut microbiota was predicted by using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). Weekly function (KEGG pathways) differences between females and males were detected by using Statistical Analysis of Metagenomic Profiles (STAMP). Results found that female pairs and male pairs were significantly different in gut microbiome diversity, compositions, and predicted metabolic profiles, importantly, females and males were also significantly dissimilar within their co-twin/triplet pairs of the mixed-sex group, infants of co-twins/triplets shared more similar features than un-related infants from different twins’ pair. Future research developing personalized interventions for vulnerable high-risk infants should consider sex, and the interaction of sex and environmental factors.
Collapse
Affiliation(s)
- Jie Chen
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Hongfei Li
- Department of Statistics, University of Connecticut, Storrs, CT, United States
| | - Sarah M Hird
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, CT, United States
| | - Wanli Xu
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Kendra Maas
- Microbial Analysis, Resources, and Services (MARS), University of Connecticut, Storrs, CT, United States
| | - Xiaomei Cong
- School of Nursing, University of Connecticut, Storrs, CT, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, CT, United States
| |
Collapse
|
153
|
Petrov ME, Jiao N, Panchanathan SS, Reifsnider E, Coonrod DV, Liu L, Krajmalnik-Brown R, Gu H, Davidson LA, Chapkin RS, Whisner CM. Protocol of the Snuggle Bug/Acurrucadito Study: a longitudinal study investigating the influences of sleep-wake patterns and gut microbiome development in infancy on rapid weight gain, an early risk factor for obesity. BMC Pediatr 2021; 21:374. [PMID: 34465311 PMCID: PMC8405858 DOI: 10.1186/s12887-021-02832-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/09/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Overweight, obesity, and associated comorbidities are a pressing global issue among children of all ages, particularly among low-income populations. Rapid weight gain (RWG) in the first 6 months of infancy contributes to childhood obesity. Suboptimal sleep-wake patterns and gut microbiota (GM) have also been associated with childhood obesity, but little is known about their influences on early infant RWG. Sleep may alter the GM and infant metabolism, and ultimately impact obesity; however, data on the interaction between sleep-wake patterns and GM development on infant growth are scarce. In this study, we aim to investigate associations of infant sleep-wake patterns and GM development with RWG at 6 months and weight gain at 12 months. We also aim to evaluate whether temporal interactions exist between infant sleep-wake patterns and GM, and if these relations influence RWG. METHODS The Snuggle Bug/ Acurrucadito study is an observational, longitudinal study investigating whether 24-h, actigraphy-assessed, sleep-wake patterns and GM development are associated with RWG among infants in their first year. Based on the Ecological Model of Growth, we propose a novel conceptual framework to incorporate sleep-wake patterns and the GM as metabolic contributors for RWG in the context of maternal-infant interactions, and familial and socio-physical environments. In total, 192 mother-infant pairs will be recruited, and sleep-wake patterns and GM development assessed at 3 and 8 weeks, and 3, 6, 9, and 12 months postpartum. Covariates including maternal and child characteristics, family and environmental factors, feeding practices and dietary intake of infants and mothers, and stool-derived metabolome and exfoliome data will be assessed. The study will apply machine learning techniques combined with logistic time-varying effect models to capture infant growth and aid in elucidating the dynamic associations between study variables and RWG. DISCUSSION Repeated, valid, and objective assessment at clinically and developmentally meaningful intervals will provide robust measures of longitudinal sleep, GM, and growth. Project findings will provide evidence for future interventions to prevent RWG in infancy and subsequent obesity. The work also may spur the development of evidence-based guidelines to address modifiable factors that influence sleep-wake and GM development and prevent childhood obesity.
Collapse
Affiliation(s)
- Megan E Petrov
- Edson College of Nursing and Health Innovation, Arizona State University, 550 N. 3rd Street, Suite 301, Phoenix, AZ, 85004, USA
| | - Nana Jiao
- Edson College of Nursing and Health Innovation, Arizona State University, 550 N. 3rd Street, Suite 301, Phoenix, AZ, 85004, USA
| | - Sarada S Panchanathan
- Valleywise Comprehensive Health Center - Phoenix (Pediatric Clinic), 2525 E. Roosevelt St., Phoenix, AZ, 85008, USA
- College of Medicine Phoenix, University of Arizona, Phoenix, AZ, 85007, USA
| | - Elizabeth Reifsnider
- Edson College of Nursing and Health Innovation, Arizona State University, 550 N. 3rd Street, Suite 301, Phoenix, AZ, 85004, USA
| | - Dean V Coonrod
- Valleywise Health, Department of Obstetrics and Gynecology, 2525 E. Roosevelt St., Phoenix, AZ, 85008, USA
| | - Li Liu
- Biodesign Institute, Arizona State University, 1001 S. McAllister Ave BDA230B, Tempe, AZ, 85287, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, 1001 S. McAllister Ave, PO Box 875701, Tempe, AZ, 85287, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, 550 N. 3rd. Street, Suite 501, Phoenix, AZ, 85004, USA
| | - Laurie A Davidson
- Department of Nutrition and Food Science, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, 2253 TAMU, 112 Cater-Mattil, College Station, TX, 77843, USA
| | - Robert S Chapkin
- Department of Nutrition and Food Science, Program in Integrative Nutrition and Complex Diseases, Texas A&M University, 2253 TAMU, 112 Cater-Mattil, College Station, TX, 77843, USA
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, 550 N. 3rd. Street, Suite 501, Phoenix, AZ, 85004, USA.
| |
Collapse
|
154
|
Nguyen QP, Karagas MR, Madan JC, Dade E, Palys TJ, Morrison HG, Pathmasiri WW, McRitche S, Sumner SJ, Frost HR, Hoen AG. Associations between the gut microbiome and metabolome in early life. BMC Microbiol 2021; 21:238. [PMID: 34454437 PMCID: PMC8400760 DOI: 10.1186/s12866-021-02282-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The infant intestinal microbiome plays an important role in metabolism and immune development with impacts on lifelong health. The linkage between the taxonomic composition of the microbiome and its metabolic phenotype is undefined and complicated by redundancies in the taxon-function relationship within microbial communities. To inform a more mechanistic understanding of the relationship between the microbiome and health, we performed an integrative statistical and machine learning-based analysis of microbe taxonomic structure and metabolic function in order to characterize the taxa-function relationship in early life. RESULTS Stool samples collected from infants enrolled in the New Hampshire Birth Cohort Study (NHBCS) at approximately 6-weeks (n = 158) and 12-months (n = 282) of age were profiled using targeted and untargeted nuclear magnetic resonance (NMR) spectroscopy as well as DNA sequencing of the V4-V5 hypervariable region from the bacterial 16S rRNA gene. There was significant inter-omic concordance based on Procrustes analysis (6 weeks: p = 0.056; 12 months: p = 0.001), however this association was no longer significant when accounting for phylogenetic relationships using generalized UniFrac distance metric (6 weeks: p = 0.376; 12 months: p = 0.069). Sparse canonical correlation analysis showed significant correlation, as well as identifying sets of microbe/metabolites driving microbiome-metabolome relatedness. Performance of machine learning models varied across different metabolites, with support vector machines (radial basis function kernel) being the consistently top ranked model. However, predictive R2 values demonstrated poor predictive performance across all models assessed (avg: - 5.06% -- 6 weeks; - 3.7% -- 12 months). Conversely, the Spearman correlation metric was higher (avg: 0.344-6 weeks; 0.265-12 months). This demonstrated that taxonomic relative abundance was not predictive of metabolite concentrations. CONCLUSIONS Our results suggest a degree of overall association between taxonomic profiles and metabolite concentrations. However, lack of predictive capacity for stool metabolic signatures reflects, in part, the possible role of functional redundancy in defining the taxa-function relationship in early life as well as the bidirectional nature of the microbiome-metabolome association. Our results provide evidence in favor of a multi-omic approach for microbiome studies, especially those focused on health outcomes.
Collapse
Affiliation(s)
- Quang P. Nguyen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Lebanon, NH USA
| | - Juliette C. Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Lebanon, NH USA
- Department of Pediatrics, Children’s Hospital at Dartmouth, Hanover, NH USA
| | - Erika Dade
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - Thomas J. Palys
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - Hilary G. Morrison
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA USA
| | - Wimal W. Pathmasiri
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Susan McRitche
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Susan J. Sumner
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - H. Robert Frost
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth College, Hanover, NH USA
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Lebanon, NH USA
| |
Collapse
|
155
|
Xiao L, Wang J, Zheng J, Li X, Zhao F. Deterministic transition of enterotypes shapes the infant gut microbiome at an early age. Genome Biol 2021; 22:243. [PMID: 34429130 PMCID: PMC8383385 DOI: 10.1186/s13059-021-02463-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/05/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The succession of the gut microbiota during the first few years plays a vital role in human development. We elucidate the characteristics and alternations of the infant gut microbiota to better understand the correlation between infant health and microbiota maturation. RESULTS We collect 13,776 fecal samples or datasets from 1956 infants between 1 and 3 years of age, based on multi-population cohorts covering 17 countries. The characteristics of the gut microbiota are analyzed based on enterotype and an ecological model. Clinical information (n = 2287) is integrated to understand outcomes of different developmental patterns. Infants whose gut microbiota are dominated by Firmicutes and Bifidobacterium exhibit typical characteristics of early developmental stages, such as unstable community structure and low microbiome maturation, while those driven by Bacteroides and Prevotella are characterized by higher diversity and stronger connections in the gut microbial community. We further reveal a geography-related pattern in global populations. Through ecological modeling and functional analysis, we demonstrate that the transition of the gut microbiota from infants towards adults follows a deterministic pattern; as infants grow up, the dominance of Firmicutes and Bifidobacterium is replaced by that of Bacteroides and Prevotella, along with shifts in specific metabolic pathways. CONCLUSIONS By leveraging the extremely large datasets and enterotype-based microbiome analysis, we decipher the colonization and transition of the gut microbiota in infants from a new perspective. We further introduce an ecological model to estimate the tendency of enterotype transitions, and demonstrated that the transition of infant gut microbiota was deterministic and predictable.
Collapse
Affiliation(s)
- Liwen Xiao
- Computational Genomics Laboratory, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinfeng Wang
- Computational Genomics Laboratory, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jiayong Zheng
- Department of Gynecology and Obstetrics, Wenzhou People's Hospital, Wenzhou, 325000, China
| | - Xiaoqing Li
- Department of Gynecology and Obstetrics, Wenzhou People's Hospital, Wenzhou, 325000, China
| | - Fangqing Zhao
- Computational Genomics Laboratory, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
156
|
Haddad EN, Comstock SS. Archive for Research in Child Health (ARCH) and Baby Gut: Study Protocol for a Remote, Prospective, Longitudinal Pregnancy and Birth Cohort to Address Microbiota Development and Child Health. Methods Protoc 2021; 4:mps4030052. [PMID: 34449678 PMCID: PMC8395764 DOI: 10.3390/mps4030052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
The infant gut microbiome is shaped by numerous factors such as diet and the maternal microbiota and is also associated with later atopy and obesity. The Archive for Research in Child Health and Baby Gut (ARCHBG) cohort was established in 2015 to (1) understand how the development of the infant gut microbiota is associated with atopy, obesity, and gastrointestinal disease and (2) characterize the associations of maternal pre-pregnancy BMI and infant diet with the development of the gut microbiota. Study participants for ARCHBG are convenience samples recruited through two pipelines in Lansing and Traverse City, Michigan: (1) Archive for Research in Child Health (ARCHGUT) and (2) BABYGUT. A total of (n = 51) mother–infant dyads have been enrolled to date. This prospective cohort study collects maternal pre-pregnancy fecal samples, maternal data, child fecal samples at four timepoints (one week, six months, 12 months, and 24 months), and child data up to five years of age. All samples and data are collected remotely by mail, phone, or drop-off at select locations. Of all participants enrolled, 76.5% (n = 39) of infants have a complete record of stool samples. At least 88.2% (n = 45) of fecal samples were submitted at each timepoint. ARCHBG will allow for a nuanced understanding of the temporal development of the infant gut microbiome and numerous child health outcomes.
Collapse
|
157
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. The role of the gut microbiome in the development of schizophrenia. Schizophr Res 2021; 234:4-23. [PMID: 32336581 DOI: 10.1016/j.schres.2020.02.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a heterogeneous neurodevelopmental disorder involving the convergence of a complex and dynamic bidirectional interaction of genetic expression and the accumulation of prenatal and postnatal environmental risk factors. The development of the neural circuitry underlying social, cognitive and emotional domains requires precise regulation from molecular signalling pathways, especially during critical periods or "windows", when the brain is particularly sensitive to the influence of environmental input signalling. Many of the brain regions involved, and the molecular substrates sub-serving these domains are responsive to life-long microbiota-gut-brain (MGB) axis signalling. This intricate microbial signalling system communicates with the brain via the vagus nerve, immune system, enteric nervous system, enteroendocrine signalling and production of microbial metabolites, such as short-chain fatty acids. Preclinical data has demonstrated that MGB axis signalling influences neurotransmission, neurogenesis, myelination, dendrite formation and blood brain barrier development, and modulates cognitive function and behaviour patterns, such as, social interaction, stress management and locomotor activity. Furthermore, preliminary clinical studies suggest altered gut microbiota profiles in schizophrenia. Unravelling MGB axis signalling in the context of an evolving dimensional framework in schizophrenia may provide a more complete understanding of the neurobiological architecture of this complex condition and offers the possibility of translational interventions.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry, Trinity College Dublin, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
158
|
Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr 2021; 12:1239-1285. [PMID: 33693453 PMCID: PMC8321864 DOI: 10.1093/advances/nmaa181] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the gut microbiota has emerged as a key component in regulating brain processes and behavior. Diet is one of the major factors involved in shaping the gut microbiota composition across the lifespan. However, whether and how diet can affect the brain via its effects on the microbiota is only now beginning to receive attention. Several mechanisms for gut-to-brain communication have been identified, including microbial metabolites, immune, neuronal, and metabolic pathways, some of which could be prone to dietary modulation. Animal studies investigating the potential of nutritional interventions on the microbiota-gut-brain axis have led to advancements in our understanding of the role of diet in this bidirectional communication. In this review, we summarize the current state of the literature triangulating diet, microbiota, and host behavior/brain processes and discuss potential underlying mechanisms. Additionally, determinants of the responsiveness to a dietary intervention and evidence for the microbiota as an underlying modulator of the effect of diet on brain health are outlined. In particular, we emphasize the understudied use of whole-dietary approaches in this endeavor and the need for greater evidence from clinical populations. While promising results are reported, additional data, specifically from clinical cohorts, are required to provide evidence-based recommendations for the development of microbiota-targeted, whole-dietary strategies to improve brain and mental health.
Collapse
Affiliation(s)
| | | | - Wolfgang Marx
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
| | - Harriet Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Felice Jacka
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Black Dog Institute, Randwick, NSW, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Timothy G Dinan
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
159
|
Murphy K, O'Donovan AN, Caplice NM, Ross RP, Stanton C. Exploring the Gut Microbiota and Cardiovascular Disease. Metabolites 2021; 11:metabo11080493. [PMID: 34436434 PMCID: PMC8401482 DOI: 10.3390/metabo11080493] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) has been classified as one of the leading causes of morbidity and mortality worldwide. CVD risk factors include smoking, hypertension, dyslipidaemia, obesity, inflammation and diabetes. The gut microbiota can influence human health through multiple interactions and community changes are associated with the development and progression of numerous disease states, including CVD. The gut microbiota are involved in the production of several metabolites, such as short-chain fatty acids (SCFAs), bile acids and trimethylamine-N-oxide (TMAO). These products of microbial metabolism are important modulatory factors and have been associated with an increased risk of CVD. Due to its association with CVD development, the gut microbiota has emerged as a target for therapeutic approaches. In this review, we summarise the current knowledge on the role of the gut microbiome in CVD development, and associated microbial communities, functions, and metabolic profiles. We also discuss CVD therapeutic interventions that target the gut microbiota such as probiotics and faecal microbiota transplantation.
Collapse
Affiliation(s)
- Kiera Murphy
- Teagasc Food Research Centre, Moorepark, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| | - Aoife N O'Donovan
- Teagasc Food Research Centre, Moorepark, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- VistaMilk SFI Research Centre, Teagasc, Moorepark, Co. Cork P61 C996, Ireland
| | - Noel M Caplice
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- Centre for Research in Vascular Biology, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- VistaMilk SFI Research Centre, Teagasc, Moorepark, Co. Cork P61 C996, Ireland
| |
Collapse
|
160
|
Phillips-Farfán B, Gómez-Chávez F, Medina-Torres EA, Vargas-Villavicencio JA, Carvajal-Aguilera K, Camacho L. Microbiota Signals during the Neonatal Period Forge Life-Long Immune Responses. Int J Mol Sci 2021; 22:ijms22158162. [PMID: 34360926 PMCID: PMC8348731 DOI: 10.3390/ijms22158162] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/27/2022] Open
Abstract
The microbiota regulates immunological development during early human life, with long-term effects on health and disease. Microbial products include short-chain fatty acids (SCFAs), formyl peptides (FPs), polysaccharide A (PSA), polyamines (PAs), sphingolipids (SLPs) and aryl hydrocarbon receptor (AhR) ligands. Anti-inflammatory SCFAs are produced by Actinobacteria, Bacteroidetes, Firmicutes, Spirochaetes and Verrucomicrobia by undigested-carbohydrate fermentation. Thus, fiber amount and type determine their occurrence. FPs bind receptors from the pattern recognition family, those from commensal bacteria induce a different response than those from pathogens. PSA is a capsular polysaccharide from B. fragilis stimulating immunoregulatory protein expression, promoting IL-2, STAT1 and STAT4 gene expression, affecting cytokine production and response modulation. PAs interact with neonatal immunity, contribute to gut maturation, modulate the gut–brain axis and regulate host immunity. SLPs are composed of a sphingoid attached to a fatty acid. Prokaryotic SLPs are mostly found in anaerobes. SLPs are involved in proliferation, apoptosis and immune regulation as signaling molecules. The AhR is a transcription factor regulating development, reproduction and metabolism. AhR binds many ligands due to its promiscuous binding site. It participates in immune tolerance, involving lymphocytes and antigen-presenting cells during early development in exposed humans.
Collapse
Affiliation(s)
- Bryan Phillips-Farfán
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Fernando Gómez-Chávez
- Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (F.G.-C.); (J.A.V.-V.)
- Cátedras CONACyT-Instituto Nacional de Pediatría, México City 04530, Mexico
- Departamento de Formación Básica Disciplinaria, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | | | | | - Karla Carvajal-Aguilera
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
| | - Luz Camacho
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, México City 04530, Mexico; (B.P.-F.); (K.C.-A.)
- Correspondence:
| |
Collapse
|
161
|
Sun S, Zhu X, Huang X, Murff HJ, Ness RM, Seidner DL, Sorgen AA, Blakley IC, Yu C, Dai Q, Azcarate-Peril MA, Shrubsole MJ, Fodor AA. On the robustness of inference of association with the gut microbiota in stool, rectal swab and mucosal tissue samples. Sci Rep 2021; 11:14828. [PMID: 34290321 PMCID: PMC8295290 DOI: 10.1038/s41598-021-94205-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota plays an important role in human health and disease. Stool, rectal swab and rectal mucosal tissue samples have been used in individual studies to survey the microbial community but the consequences of using these different sample types are not completely understood. In this study, we report differences in stool, rectal swab and rectal mucosal tissue microbial communities with shotgun metagenome sequencing of 1397 stool, swab and mucosal tissue samples from 240 participants. The taxonomic composition of stool and swab samples was distinct, but less different to each other than mucosal tissue samples. Functional profile differences between stool and swab samples are smaller, but mucosal tissue samples remained distinct from the other two types. When the taxonomic and functional profiles were used for inference in association with host phenotypes of age, sex, body mass index (BMI), antibiotics or non-steroidal anti-inflammatory drugs (NSAIDs) use, hypothesis testing using either stool or rectal swab gave broadly significantly correlated results, but inference performed on mucosal tissue samples gave results that were generally less consistent with either stool or swab. Our study represents an important resource for determination of how inference can change for taxa and pathways depending on the choice of where to sample within the human gut.
Collapse
Affiliation(s)
- Shan Sun
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Xiangzhu Zhu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiang Huang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Harvey J Murff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Reid M Ness
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas L Seidner
- Digestive Disease and Surgical Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alicia A Sorgen
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Ivory C Blakley
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qi Dai
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine and Microbiome Core Facility, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Martha J Shrubsole
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anthony A Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, 9331 Robert D. Snyder Rd, Room 361, Charlotte, NC, 28223, USA.
| |
Collapse
|
162
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
163
|
Lay C, Chu CW, Purbojati RW, Acerbi E, Drautz-Moses DI, de Sessions PF, Jie S, Ho E, Kok YJ, Bi X, Chen S, Mak SY, Chua MC, Goh AEN, Chiang WC, Rao R, Chaithongwongwatthana S, Khemapech N, Chongsrisawat V, Martin R, Roeselers G, Ho YS, Hibberd ML, Schuster SC, Knol J. A synbiotic intervention modulates meta-omics signatures of gut redox potential and acidity in elective caesarean born infants. BMC Microbiol 2021; 21:191. [PMID: 34172012 PMCID: PMC8229302 DOI: 10.1186/s12866-021-02230-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The compromised gut microbiome that results from C-section birth has been hypothesized as a risk factor for the development of non-communicable diseases (NCD). In a double-blind randomized controlled study, 153 infants born by elective C-section received an infant formula supplemented with either synbiotic, prebiotics, or unsupplemented from birth until 4 months old. Vaginally born infants were included as a reference group. Stool samples were collected from day 3 till week 22. Multi-omics were deployed to investigate the impact of mode of delivery and nutrition on the development of the infant gut microbiome, and uncover putative biological mechanisms underlying the role of a compromised microbiome as a risk factor for NCD. RESULTS As early as day 3, infants born vaginally presented a hypoxic and acidic gut environment characterized by an enrichment of strict anaerobes (Bifidobacteriaceae). Infants born by C-section presented the hallmark of a compromised microbiome driven by an enrichment of Enterobacteriaceae. This was associated with meta-omics signatures characteristic of a microbiome adapted to a more oxygen-rich gut environment, enriched with genes associated with reactive oxygen species metabolism and lipopolysaccharide biosynthesis, and depleted in genes involved in the metabolism of milk carbohydrates. The synbiotic formula modulated expression of microbial genes involved in (oligo)saccharide metabolism, which emulates the eco-physiological gut environment observed in vaginally born infants. The resulting hypoxic and acidic milieu prevented the establishment of a compromised microbiome. CONCLUSIONS This study deciphers the putative functional hallmarks of a compromised microbiome acquired during C-section birth, and the impact of nutrition that may counteract disturbed microbiome development. TRIAL REGISTRATION The study was registered in the Dutch Trial Register (Number: 2838 ) on 4th April 2011.
Collapse
Affiliation(s)
| | | | - Rikky Wenang Purbojati
- Singapore Centre For Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| | - Enzo Acerbi
- Danone Nutricia Research, Singapore, Singapore
| | - Daniela I Drautz-Moses
- Singapore Centre For Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| | | | - Song Jie
- Genome Institute of Singapore, Singapore, Singapore
| | - Eliza Ho
- Genome Institute of Singapore, Singapore, Singapore
| | - Yee Jiun Kok
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Shuwen Chen
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Mei Chien Chua
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Anne E N Goh
- KK Women's and Children's Hospital, Singapore, Singapore
| | | | - Rajeshwar Rao
- KK Women's and Children's Hospital, Singapore, Singapore
| | | | - Nipon Khemapech
- King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Voranush Chongsrisawat
- King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rocio Martin
- Danone Nutricia Research, Utrecht, The Netherlands
| | | | - Ying Swan Ho
- Bioprocessing Technology Institute, Singapore, Singapore
| | - Martin L Hibberd
- Genome Institute of Singapore, Singapore, Singapore
- London School of Hygiene and Tropical Medicine, London, UK
| | - Stephan C Schuster
- Singapore Centre For Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| | - Jan Knol
- Danone Nutricia Research, Utrecht, The Netherlands.
- Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
164
|
Patangia DV, Ryan CA, Dempsey E, Stanton C, Ross RP. Vertical transfer of antibiotics and antibiotic resistant strains across the mother/baby axis. Trends Microbiol 2021; 30:47-56. [PMID: 34172345 DOI: 10.1016/j.tim.2021.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/06/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
Antibiotic resistance is a health and socioeconomic crisis recognized as a serious threat affecting humans worldwide. Overuse of antibiotics enhances the spread of multidrug-resistant bacteria, causing drug-resistant infections which can be difficult to treat. This resistance, mostly of the acquired type, is thus a major clinical issue. Acquired resistance can occur by horizontal transfer of genes between bacteria (community settings), by vertical transmission that can occur between mother and her offspring at birth and during lactation, or spontaneously due to antibiotic exposure. While there have been multiple studies about the horizontal transfer of antibiotic-resistance genes, not many studies have been conducted to study their vertical transmission. Vertical transmission is of importance as the early bacterial colonization of infants has an impact on their health and immune programming throughout life. This review discusses some possible mechanisms of mother-to-infant transmission of antibiotics and antibiotic-resistant strains and addresses the knowledge gaps for further studies.
Collapse
Affiliation(s)
- Dhrati V Patangia
- School of Microbiology, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - C Anthony Ryan
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland; APC Microbiome Ireland, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, Cork, Ireland.
| |
Collapse
|
165
|
Nutritional and therapeutic approaches for protecting human gut microbiota from psychotropic treatments. Prog Neuropsychopharmacol Biol Psychiatry 2021; 108:110182. [PMID: 33232785 DOI: 10.1016/j.pnpbp.2020.110182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Emerging evidence highlighted the essential role played by the microbiota-gut-brain axis in maintaining human homeostasis, including nutrition, immunity, and metabolism. Much recent work has linked the gut microbiota to many psychiatric and neurodegenerative disorders such as depression, schizophrenia, and Alzheimer's disease. Shared gut microbiota alterations or dysbiotic microbiota have been identified in these separate disorders relative to controls. Much attention has focused on the bidirectional interplay between the gut microbiota and the brain, establishing gut dysbiotic status as a critical factor in psychiatric disorders. Still, the antibiotic-like effect of psychotropic drugs, medications used for the treatment of these disorders, on gut microbiota is largely neglected. In this review, we summarize the current findings on the impact of psychotropics on gut microbiota and how their antimicrobial potency can trigger dysbiosis. We also discuss the potential therapeutic strategies, including probiotics, prebiotics, and fecal transplantation, to attenuate the dysbiosis related to psychotropics intake.
Collapse
|
166
|
Wagner C, Torow N, Hornef MW, Lelouard H. Spatial and temporal key steps in early-life intestinal immune system development and education. FEBS J 2021; 289:4731-4757. [PMID: 34076962 DOI: 10.1111/febs.16047] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Education of our intestinal immune system early in life strongly influences adult health. This education strongly relies on series of events that must occur in well-defined time windows. From initial colonization by maternal-derived microbiota during delivery to dietary changes from mother's milk to solid foods at weaning, these early-life events have indeed long-standing consequences on our immunity, facilitating tolerance to environmental exposures or, on the contrary, increasing the risk of developing noncommunicable diseases such as allergies, asthma, obesity, and inflammatory bowel diseases. In this review, we provide an outline of the recent advances in our understanding of these events and how they are mechanistically related to intestinal immunity development and education. First, we review the susceptibility of neonates to infections and inflammatory diseases, related to their immune system and microbiota changes. Then, we highlight the maternal factors involved in protection and education of the mucosal immune system of the offspring, the role of the microbiota, and the nature of neonatal immune system until weaning. We also present how the development of some immune responses is intertwined in temporal and spatial windows of opportunity. Finally, we discuss pending questions regarding the neonate particular immune status and the activation of the intestinal immune system at weaning.
Collapse
Affiliation(s)
- Camille Wagner
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Natalia Torow
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | | |
Collapse
|
167
|
Alabduljabbar S, Zaidan SA, Lakshmanan AP, Terranegra A. Personalized Nutrition Approach in Pregnancy and Early Life to Tackle Childhood and Adult Non-Communicable Diseases. Life (Basel) 2021; 11:life11060467. [PMID: 34073649 PMCID: PMC8224671 DOI: 10.3390/life11060467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
The development of childhood and adult non-communicable diseases (NCD) is associated with environmental factors, starting from intrauterine life. A new theory finds the roots of epigenetic programming in parental gametogenesis, continuing during embryo development, fetal life, and finally in post-natal life. Maternal health status and poor nutrition are widely recognized as implications in the onset of childhood and adult diseases. Early nutrition, particularly breastfeeding, also plays a primary role in affecting the health status of an individual later in life. A poor maternal diet during pregnancy and lack of breastfeeding can cause a nutrient deficiency that affects the gut microbiota, and acts as a cofactor for many pathways, impacting the epigenetic controls and transcription of genes involved in the metabolism, angiogenesis, and other pathways, leading to NCDs in adult life. Both maternal and fetal genetic backgrounds also affect nutrient adsorption and functioning at the cellular level. This review discusses the most recent evidence on maternal nutrition and breastfeeding in the development of NCD, the potentiality of the omics technologies in uncovering the molecular mechanisms underlying it, with the future prospective of applying a personalized nutrition approach to prevent and treat NCD from the beginning of fetal life.
Collapse
|
168
|
Sabater C, Ruiz L, Margolles A. A Machine Learning Approach to Study Glycosidase Activities from Bifidobacterium. Microorganisms 2021; 9:1034. [PMID: 34064844 PMCID: PMC8151561 DOI: 10.3390/microorganisms9051034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
This study aimed to recover metagenome-assembled genomes (MAGs) from human fecal samples to characterize the glycosidase profiles of Bifidobacterium species exposed to different prebiotic oligosaccharides (galacto-oligosaccharides, fructo-oligosaccharides and human milk oligosaccharides, HMOs) as well as high-fiber diets. A total of 1806 MAGs were recovered from 487 infant and adult metagenomes. Unsupervised and supervised classification of glycosidases codified in MAGs using machine-learning algorithms allowed establishing characteristic hydrolytic profiles for B. adolescentis, B. bifidum, B. breve, B. longum and B. pseudocatenulatum, yielding classification rates above 90%. Glycosidase families GH5 44, GH32, and GH110 were characteristic of B. bifidum. The presence or absence of GH1, GH2, GH5 and GH20 was characteristic of B. adolescentis, B. breve and B. pseudocatenulatum, while families GH1 and GH30 were relevant in MAGs from B. longum. These characteristic profiles allowed discriminating bifidobacteria regardless of prebiotic exposure. Correlation analysis of glycosidase activities suggests strong associations between glycosidase families comprising HMOs-degrading enzymes, which are often found in MAGs from the same species. Mathematical models here proposed may contribute to a better understanding of the carbohydrate metabolism of some common bifidobacteria species and could be extrapolated to other microorganisms of interest in future studies.
Collapse
Affiliation(s)
- Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Paseo Río Linares S/N, 33300 Villaviciosa, Asturias, Spain; (L.R.); (A.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| |
Collapse
|
169
|
Morais J, Marques C, Faria A, Teixeira D, Barreiros-Mota I, Durão C, Araújo J, Ismael S, Brito S, Cardoso M, Macedo I, Pereira E, Tomé T, Calhau C. Influence of Human Milk on Very Preterms' Gut Microbiota and Alkaline Phosphatase Activity. Nutrients 2021; 13:1564. [PMID: 34066473 PMCID: PMC8148101 DOI: 10.3390/nu13051564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 01/12/2023] Open
Abstract
The FEEDMI Study (NCT03663556) evaluated the influence of infant feeding (mother's own milk (MOM), donor human milk (DHM) and formula) on the fecal microbiota composition and alkaline phosphatase (ALP) activity in extremely and very preterm infants (≤32 gestational weeks). In this observational study, preterm infants were recruited within the first 24 h after birth. Meconium and fecal samples were collected at four time points (between the 2nd and the 26th postnatal days. Fecal microbiota was analyzed by RT-PCR and by 16S rRNA sequencing. Fecal ALP activity, a proposed specific biomarker of necrotizing enterocolitis (NEC), was evaluated by spectrophotometry at the 26th postnatal day. A total of 389 fecal samples were analyzed from 117 very preterm neonates. Human milk was positively associated with beneficial bacteria, such as Bifidobacterium, Bacteroides ovatus, and Akkermancia muciniphila, as well as bacterial richness. Neonates fed with human milk during the first week of life had increased Bifidobacterium content and fecal ALP activity on the 26th postnatal day. These findings point out the importance of MOM and DHM in the establishment of fecal microbiota on neonates prematurely delivered. Moreover, these results suggest an ALP pathway by which human milk may protect against NEC.
Collapse
Affiliation(s)
- Juliana Morais
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Cláudia Marques
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Ana Faria
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Diana Teixeira
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Medical School, Unidade Universitária Lifestyle Medicine José de Mello Saúde, 1169-056 Lisboa, Portugal
| | - Inês Barreiros-Mota
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Catarina Durão
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- EPIUnit-Institute of Public Health, Universidade do Porto, 4050-600 Porto, Portugal
| | - João Araújo
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Shámila Ismael
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CHRC-Comprehensive Health Research Centre, CEDOC-Chronic Diseases Research Center, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Sara Brito
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Manuela Cardoso
- Nutrition and Dietetics Unit, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal;
| | - Israel Macedo
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Esmeralda Pereira
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Teresa Tomé
- Pediatrics Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, 2890-495 Lisboa, Portugal; (S.B.); (I.M.); (E.P.); (T.T.)
| | - Conceição Calhau
- Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (J.M.); (C.M.); (A.F.); (D.T.); (I.B.-M.); (C.D.); (J.A.); (S.I.)
- CINTESIS-Center for Health Technology Services Research, Faculdade de Ciências Médicas|NOVA Medical School, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
- NOVA Medical School, Unidade Universitária Lifestyle Medicine José de Mello Saúde, 1169-056 Lisboa, Portugal
| |
Collapse
|
170
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
171
|
Gestational age-dependent development of the neonatal metabolome. Pediatr Res 2021; 89:1396-1404. [PMID: 32942288 DOI: 10.1038/s41390-020-01149-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/08/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prematurity is a severe pathophysiological condition, however, little is known about the gestational age-dependent development of the neonatal metabolome. METHODS Using an untargeted liquid chromatography-tandem mass spectrometry metabolomics protocol, we measured over 9000 metabolites in 298 neonatal residual heel prick dried blood spots retrieved from the Danish Neonatal Screening Biobank. By combining multiple state-of-the-art metabolome mining tools, we retrieved chemical structural information at a broad level for over 5000 (60%) metabolites and assessed their relation to gestational age. RESULTS A total of 1459 (~16%) metabolites were significantly correlated with gestational age (false discovery rate-adjusted P < 0.05), whereas 83 metabolites explained on average 48% of the variance in gestational age. Using a custom algorithm based on hypergeometric testing, we identified compound classes (617 metabolites) overrepresented with metabolites correlating with gestational age (P < 0.05). Metabolites significantly related to gestational age included bile acids, carnitines, polyamines, amino acid-derived compounds, nucleotides, phosphatidylcholines and dipeptides, as well as treatment-related metabolites, such as antibiotics and caffeine. CONCLUSIONS Our findings elucidate the gestational age-dependent development of the neonatal blood metabolome and suggest that the application of metabolomics tools has great potential to reveal novel biochemical underpinnings of disease and improve our understanding of complex pathophysiological mechanisms underlying prematurity-associated disorders. IMPACT A large variation in the neonatal dried blood spot metabolome from residual heel pricks stored at the Danish Neonatal Screening Biobank can be explained by gestational age. While previous studies have assessed the relation of selected metabolic markers to gestational age, this study assesses metabolome-wide changes related to prematurity. Using a combination of recently developed metabolome mining tools, we assess the relation of over 9000 metabolic features to gestational age. The ability to assess metabolome-wide changes related to prematurity in neonates could pave the way to finding novel biochemical underpinnings of health complications related to preterm birth.
Collapse
|
172
|
Gut bacterial profile in Indian children of varying nutritional status: a comparative pilot study. Eur J Nutr 2021; 60:3971-3985. [PMID: 33929588 PMCID: PMC8085102 DOI: 10.1007/s00394-021-02571-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Childhood malnutrition is a multifactorial disease, responsible for nearly half of all deaths in children under five. Lately, the probable association of a dysbiotic gut to malnutrition is also being eagerly investigated. The current study is an attempt to investigate this purported association through assessing the abundance of major gut bacterial phyla (Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria), probionts (Bifidobacteria and Lactobacillus), butyrogens (Faecalibacterium and Roseburia) and pathogens (Escherichia and Klebsiella). METHODS The study was conducted in the suburbs of Chandigarh, India in the year 2017. The children enrolled in the study were part of Anganwadis (Rural Child Care Centres) set up under Integrated Child Development Scheme (ICDS) of Government of India where community-based management approach is being widely used for treatment of malnutrition. We used qPCR based absolute quantification as well as the 16S rRNA amplicon sequencing approach for our study. The study population included 30 children in the age group of 2-5 years who were categorized into three groups Healthy, Moderate Acute Malnutrition (MAM) and Severe Acute Malnutrition (SAM), with 10 children in each group. The selection of participants was made based on Z scores. Further, statistical tools like the One-way ANOVA, PCA and PLSDA were employed to analyze and compare the gut bacterial profile. RESULTS Our investigation through the qPCR (Absolute quantification) approach revealed a significantly higher abundance of Actinobacteria in healthy, in comparison to children suffering from Severe Acute Malnutrition (SAM). Consequently, the same trend was also reflected with respect to Bifidobacterium, a prominent member of the Actinobacteria phylum. Conversely, a significant higher abundance of Lactobacillus with the diminishing nutritional status was recorded. Escherichia showed a significant higher abundance in healthy subjects compared to the malnourished; however, no such difference in abundance of Klebsiella was observed. The other target phyla [Bacteroidetes, Firmicutes and Proteobacteria] and genera (Faecalibacterium and Roseburia) showed differences in abundance; however, these were non-significant. Similarly, the bacterial taxonomy analysis of 16S rRNA gene amplicon sequencing data revealed the higher abundance of phylum Actinobacteria and its member Bifidobacterium with lower prevalence of Lactobacillus in healthy children. CONCLUSION The pattern of gut microbiota profile in malnourished subjects suggests a dysbiotic gut depleted in Bifidobacteria, a core member of the consortia of beneficial anaerobes of the healthy child gut.
Collapse
|
173
|
Taboada B, Morán P, Serrano-Vázquez A, Iša P, Rojas-Velázquez L, Pérez-Juárez H, López S, Torres J, Ximenez C, Arias CF. The gut virome of healthy children during the first year of life is diverse and dynamic. PLoS One 2021; 16:e0240958. [PMID: 33852569 PMCID: PMC8046192 DOI: 10.1371/journal.pone.0240958] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/30/2021] [Indexed: 01/21/2023] Open
Abstract
In this work, we determined the diversity and dynamics of the gut virome of infants during the first year of life. Fecal samples were collected monthly, from birth to one year of age, from three healthy children living in a semi-rural village in Mexico. Most of the viral reads were classified into six families of bacteriophages including five dsDNA virus families of the order Caudovirales, with Siphoviridae and Podoviridae being the most abundant. Eukaryotic viruses were detected as early as two weeks after birth and remained present all along the first year of life. Thirty-four different eukaryotic virus families were found, where eight of these families accounted for 98% of all eukaryotic viral reads: Anelloviridae, Astroviridae, Caliciviridae, Genomoviridae, Parvoviridae, Picornaviridae, Reoviridae and the plant-infecting viruses of the Virgaviridae family. Some viruses in these families are known human pathogens, and it is surprising that they were found during the first year of life in infants without gastrointestinal symptoms. The eukaryotic virus species richness found in this work was higher than that observed in previous studies; on average between 7 and 24 virus species were identified per sample. The richness and abundance of the eukaryotic virome significantly increased during the second semester of life, probably because of an increased environmental exposure of infants with age. Our findings suggest an early and permanent contact of infants with a diverse array of bacteriophages and eukaryotic viruses, whose composition changes over time. The bacteriophages and eukaryotic viruses found in these children could represent a metastable virome, whose potential influence on the development of the infant's immune system or on the health of the infants later in life, remains to be investigated.
Collapse
Affiliation(s)
- Blanca Taboada
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Patricia Morán
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Pavel Iša
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Susana López
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
- * E-mail: (CFA); (CX); (JT)
| | - Cecilia Ximenez
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
- * E-mail: (CFA); (CX); (JT)
| | - Carlos F. Arias
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- * E-mail: (CFA); (CX); (JT)
| |
Collapse
|
174
|
Shaterian N, Abdi F, Ghavidel N, Alidost F. Role of cesarean section in the development of neonatal gut microbiota: A systematic review. Open Med (Wars) 2021; 16:624-639. [PMID: 33869784 PMCID: PMC8035494 DOI: 10.1515/med-2021-0270] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background The delivery mode is one of the factors affecting the type of colonization of the human gut. Gut colonization affects all stages of the human life cycle, and the type of gut microbiome can contribute to immune system function, the development of some diseases, and brain development; and it has a significant impact on a newborn’s growth and development. Methods Terms defined as MeSH keywords were searched by the databases, and web search engines such as PubMed, ClinicalTrials.gov, Embase, Scopus, ProQuest, Web of Science, and Google Scholar were searched between 2010 and 2020. The quality of each study was assessed according to the Newcastle–Ottawa scale, and seven eligible and high-quality studies were analyzed. Finding The abundances of Bacteroides and Bifidobacterium during the first 3 months of life; Lactobacillus and Bacteroides during the second 3 months of life; Bacteroides and Bifidobacterium during the second 6 months of life; and Bacteroides, Enterobacter, and Streptococcus after the first year of life were higher in vaginal delivery-born infants. While infants born by cesarean section (CS) had higher abundances of Clostridium and Lactobacillus during the first 3 months of life, Enterococcus and Clostridium during the second 3 months of life, and Lactobacillus and Staphylococcus after the first year of life. Discussion Delivery mode can affect the type of the human intestinal microbiota. The CS-born babies had lower colonization rates of Bifidobacterium and Bacteroides, but they had higher colonization rates of Clostridium, Lactobacillus, Enterobacter, Enterococcus, and Staphylococcus. Given the effect of microbiota colonization on neonatal health, it is therefore recommended to conduct further studies in order to investigate the effect of the colonization on the delivery mode and on baby’s growth and development. Application to practice The aim of this study was to investigate the role of CS in the development of the neonatal gut microbiota.
Collapse
Affiliation(s)
- Negin Shaterian
- Student Research Committee, Nursing and Midwifery faculty, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abdi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Nooshin Ghavidel
- Social Determinants of Health Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farzane Alidost
- Student Research Committee, School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
175
|
Princisval L, Rebelo F, Williams BL, Coimbra AC, Crovesy L, Ferreira AL, Kac G. Association Between the Mode of Delivery and Infant Gut Microbiota Composition Up to 6 Months of Age: A Systematic Literature Review Considering the Role of Breastfeeding. Nutr Rev 2021; 80:113-127. [PMID: 33837424 DOI: 10.1093/nutrit/nuab008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
CONTEXT Cesarean section (CS), breastfeeding, and geographic location can influence the infant microbiota. OBJECTIVE In this systematic review, evidence of the association between mode of delivery and infant gut microbiota up to 6 months of age was evaluated, as was the role of breastfeeding in this association, according to PRISMA guidelines. DATA SOURCE The Pubmed, Web of Science, Scopus, Embase, Medical Database, and Open Grey databases were searched. DATA EXTRACTION A total of 31 observational studies with ≥2 infant stool collections up to the sixth month of age and a comparison of gut microbiota between CS and vaginal delivery (VD) were included. DATA ANALYSIS Infants born by CS had a lower abundance of Bifidobacterium and Bacteroides spp. at almost all points up to age 6 months. Populations of Lactobacillus, Bifidobacterium longum, Bifidobacterium catenulatum, and Escherichia coli were reduced in infants delivered by CS. Infants born by CS and exclusively breastfed had greater similarity with the microbiota of infants born by VD. CONCLUSIONS Species of Bifidobacterium and Bacteroides are potentially reduced in infants born by CS. Geographic location influenced bacterial colonization. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. 42017071285.
Collapse
Affiliation(s)
- Luciana Princisval
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Fernanda Rebelo
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Brent L Williams
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Anna Carolina Coimbra
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Louise Crovesy
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Ana Lorena Ferreira
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| | - Gilberto Kac
- Affiliation: L. Princisval, A.C. Coimbra, L. Crovesy, A.L. Ferreira, and G. Kac are with the Department of Social and Applied Nutrition, Federal University of Rio de Janeiro, Josué de Castro Nutrition Institute, Rio de Janeiro, RJ Brazil. B.L. Williams is with the Department of Epidemiology, Columbia University, Center for Infection and Immunity, New York, NY, USA. F. Rebelo is with the Oswaldo Cruz Foundation, National Institute of Women, Children and Adolescents Health Fernandes Figueira, Clinical Research Unit, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
176
|
Coker MO, Laue HE, Hoen AG, Hilliard M, Dade E, Li Z, Palys T, Morrison HG, Baker E, Karagas MR, Madan JC. Infant Feeding Alters the Longitudinal Impact of Birth Mode on the Development of the Gut Microbiota in the First Year of Life. Front Microbiol 2021; 12:642197. [PMID: 33897650 PMCID: PMC8059768 DOI: 10.3389/fmicb.2021.642197] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Cesarean-delivered (CD) infants harbor a distinct gut microbiome from vaginally delivered (VD) infants, however, during infancy, the most important driver of infant gut microbial colonization is infant feeding. Earlier studies have shown that breastfeeding is associated with higher levels of health-promoting bacteria such and Bifidobacterium and Bacteroides via modulation of the immune system, and production of metabolites. As the infant gut matures and solid foods are introduced, it is unclear whether longer duration of breast feeding restore loss of beneficial taxa within the intestinal microbiota of operatively delivered infants. Within the New Hampshire Birth Cohort Study, we evaluated the longitudinal effect of delivery mode and infant feeding on the taxonomic composition and functional capacity of developing gut microbiota in the First year of life. Microbiota of 500 stool samples collected between 6 weeks and 12 months of age (from 229 infants) were characterized by 16S ribosomal RNA sequencing. Shotgun metagenomic sequencing was also performed on 350 samples collected at either 6 weeks or 12 months of age. Among infant participants, 28% were cesarean-delivered (CD) infants and most (95%) initiated breastfeeding within the first six months of life, with 26% exclusively breastfed and 69% mixed-fed (breast milk and formula), in addition to complementary foods by age 1. Alpha (within-sample) diversity was significantly lower in CD infants compared to vaginally delivered (VD) infants (P < 0.05) throughout the study period. Bacterial community composition clustering by both delivery mode and feeding duration at 1 year of age revealed that CD infants who were breast fed for < 6 months were more dissimilar to VD infants than CD infants who breast fed for ≥ 6 months. We observed that breastfeeding modified the longitudinal impact of delivery mode on the taxonomic composition of the microbiota by 1 year of age, with an observed increase in abundance of Bacteroides fragilis and Lactobacillus with longer duration of breastfeeding among CD infants while there was an increase in Faecalibacterium for VD infants. Our findings confirm that duration of breastfeeding plays a critical role in restoring a health-promoting microbiome, call for further investigations regarding the association between breast milk exposure and health outcomes in early life.
Collapse
Affiliation(s)
- Modupe O. Coker
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- School of Dental Medicine, School of Public Health at Rutgers, Newark, NJ, United States
| | - Hannah E. Laue
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Anne G. Hoen
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Margaret Hilliard
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Erika Dade
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Zhigang Li
- Department of Biostatistics, University of Florida, Gainsville, FL, United States
| | - Thomas Palys
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Hilary G. Morrison
- Marine Biological Laboratory, Josephine Bay Paul Center, Woods Hole, MA, United States
| | - Emily Baker
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| | - Margaret R. Karagas
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
| | - Juliette C. Madan
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, United States
- Children’s Environmental Health & Disease Prevention Research Center at Dartmouth, Hanover, NH, United States
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
177
|
Investigating colonization patterns of the infant gut microbiome during the introduction of solid food and weaning from breastmilk: A cohort study protocol. PLoS One 2021; 16:e0248924. [PMID: 33798237 PMCID: PMC8018627 DOI: 10.1371/journal.pone.0248924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
The first exposures to microbes occur during infancy and it is suggested that this initial colonization influences the adult microbiota composition. Despite the important role that the gut microbiome may have in health outcomes later in life, the factors that influence its development during infancy and early childhood have not been characterized fully. Guidelines about the introduction of solid foods and cessation of breastfeeding, which is thought to have a significant role in the transition to a more adult-like microbiota, are not based on microbiome research. There is even less understanding of approaches used to transition to solid food in the preterm population. The purpose of this study is to identify the impact of early life dietary events on gut microbiome community structures and function among infants born at term and pre-term. We plan to prospectively monitor the gut microbiome of infants during two critical timepoints in microbial development: the introduction of solid foods and cessation from breastmilk. A total of 35 participants from three primary observational birth cohorts (two full-term cohorts and one pre-term cohort) will be enrolled in this sub-study. Participants will be asked to collect stool samples and fill out a study diary before, during and after the introduction of solids and again during weaning from breastmilk. We will use frequent fecal sampling analyzed using 16S rRNA gene profiling, metagenomics, metabolomics, and targeted bacterial culturing to identify and characterize the microbial communities, as well as provide insight into the phenotypic characteristics and functional capabilities of the microbes present during these transitional periods of infancy. This study will provide a comprehensive approach to detailing the effects of dietary transition from breastmilk to a more adult-like solid food diet on the microbiome and in doing so will contribute to evidence-based infant nutrition guidance.
Collapse
|
178
|
Maiuolo J, Gliozzi M, Musolino V, Carresi C, Scarano F, Nucera S, Scicchitano M, Oppedisano F, Bosco F, Ruga S, Zito MC, Macri R, Palma E, Muscoli C, Mollace V. The Contribution of Gut Microbiota-Brain Axis in the Development of Brain Disorders. Front Neurosci 2021; 15:616883. [PMID: 33833660 PMCID: PMC8021727 DOI: 10.3389/fnins.2021.616883] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
Different bacterial families colonize most mucosal tissues in the human organism such as the skin, mouth, vagina, respiratory, and gastrointestinal districts. In particular, the mammalian intestine hosts a microbial community of between 1,000 and 1,500 bacterial species, collectively called "microbiota." Co-metabolism between the microbiota and the host system is generated and the symbiotic relationship is mutually beneficial. The balance that is achieved between the microbiota and the host organism is fundamental to the organization of the immune system. Scientific studies have highlighted a direct correlation between the intestinal microbiota and the brain, establishing the existence of the gut microbiota-brain axis. Based on this theory, the microbiota acts on the development, physiology, and cognitive functions of the brain, although the mechanisms involved have not yet been fully interpreted. Similarly, a close relationship between alteration of the intestinal microbiota and the onset of several neurological pathologies has been highlighted. This review aims to point out current knowledge as can be found in literature regarding the connection between intestinal dysbiosis and the onset of particular neurological pathologies such as anxiety and depression, autism spectrum disorder, and multiple sclerosis. These disorders have always been considered to be a consequence of neuronal alteration, but in this review, we hypothesize that these alterations may be non-neuronal in origin, and consider the idea that the composition of the microbiota could be directly involved. In this direction, the following two key points will be highlighted: (1) the direct cross-talk that comes about between neurons and gut microbiota, and (2) the degree of impact of the microbiota on the brain. Could we consider the microbiota a valuable target for reducing or modulating the incidence of certain neurological diseases?
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Maria Caterina Zito
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Ernesto Palma
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Carolina Muscoli
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
- IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
179
|
Klymiuk I, Singer G, Castellani C, Trajanoski S, Obermüller B, Till H. Characterization of the Luminal and Mucosa-Associated Microbiome along the Gastrointestinal Tract: Results from Surgically Treated Preterm Infants and a Murine Model. Nutrients 2021; 13:nu13031030. [PMID: 33806771 PMCID: PMC8004827 DOI: 10.3390/nu13031030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Environmental factors, including nutritional habits or birth mode, are known key determinants for intestinal microbial composition. Investigations of the intestinal microbiome in different species in a multiplicity of studies during recent decades have revealed differential microbial patterns and quantities along the gastrointestinal (GI) tract. Characterization of the microbial pattern in various aspects is a prerequisite for nutritional interventions. In this 16S rRNA amplicon-based approach, we present a characterization of the mucosa-associated microbiome in comparison with the luminal community of four infants at the time of the closure of ileostomies and perform a systematic characterization of the corresponding luminal and mucosal microbiome from jejunal, ileal and colonic regions, as well as collected feces in mice. The most dominant taxa in infant-derived samples altered due to individual differences, and in the mucosa, Enterococcus, Clostridiumsensustricto1, Veillonella, Streptococcus and Staphylococcus were the most abundant. Two less abundant taxa differed significantly between the mucosa and lumen. In murine samples, relative abundances differed significantly, mainly between the intestinal regions. Significant differences between mouse mucosa- and lumen-derived samples could be found in the observed species with a trend to lower estimated diversity in mucosa-derived samples, as well as in the relative abundance of individual taxa. In this study, we examined the difference between the mucosal and luminal bacterial colonization of the gastrointestinal tract in a small sample cohort of preterm infants. Individual differences were characterized and statistical significance was reached in two taxa (Cupriavidus, Ralstonia). The corresponding study on the different murine intestinal regions along the GI tract showed differences all over the intestinal region.
Collapse
Affiliation(s)
- Ingeborg Klymiuk
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria; (I.K.); (S.T.)
| | - Georg Singer
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8010 Graz, Austria; (C.C.); (B.O.); (H.T.)
- Correspondence: ; Tel.: +43-316-385-83722
| | - Christoph Castellani
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8010 Graz, Austria; (C.C.); (B.O.); (H.T.)
| | - Slave Trajanoski
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria; (I.K.); (S.T.)
| | - Beate Obermüller
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8010 Graz, Austria; (C.C.); (B.O.); (H.T.)
| | - Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8010 Graz, Austria; (C.C.); (B.O.); (H.T.)
| |
Collapse
|
180
|
Administration of β-lactam antibiotics and delivery method correlate with intestinal abundances of Bifidobacteria and Bacteroides in early infancy, in Japan. Sci Rep 2021; 11:6231. [PMID: 33737648 PMCID: PMC7973812 DOI: 10.1038/s41598-021-85670-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
The intestinal microbiome changes dynamically in early infancy. Colonisation by Bifidobacterium and Bacteroides and development of intestinal immunity is interconnected. We performed a prospective observational cohort study to determine the influence of antibiotics taken by the mother immediately before delivery on the intestinal microbiome of 130 healthy Japanese infants. Faecal samples (383) were collected at 1, 3, and 6 months and analysed using next-generation sequencing. Cefazolin was administered before caesarean sections, whereas ampicillin was administered in cases with premature rupture of the membranes and in Group B Streptococcus-positive cases. Bifidobacterium and Bacteroides were dominant (60–70% mean combined occupancy) at all ages. A low abundance of Bifidobacterium was observed in infants exposed to antibiotics at delivery and at 1 and 3 months, with no difference between delivery methods. A lower abundance of Bacteroides was observed after caesarean section than vaginal delivery, irrespective of antibiotic exposure. Additionally, occupancy by Bifidobacterium at 1 and 3 months and by Bacteroides at 3 months differed between infants with and without siblings. All these differences disappeared at 6 months. Infants exposed to intrapartum antibiotics displayed altered Bifidobacterium abundance, whereas abundance of Bacteroides was largely associated with the delivery method. Existence of siblings also significantly influenced the microbiota composition of infants.
Collapse
|
181
|
Wang S, Egan M, Ryan CA, Boyaval P, Dempsey EM, Ross RP, Stanton C. A good start in life is important-perinatal factors dictate early microbiota development and longer term maturation. FEMS Microbiol Rev 2021; 44:763-781. [PMID: 32821932 PMCID: PMC7685781 DOI: 10.1093/femsre/fuaa030] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal health status is vital for the development of the offspring of humans, including physiological health and psychological functions. The complex and diverse microbial ecosystem residing within humans contributes critically to these intergenerational impacts. Perinatal factors, including maternal nutrition, antibiotic use and maternal stress, alter the maternal gut microbiota during pregnancy, which can be transmitted to the offspring. In addition, gestational age at birth and mode of delivery are indicated frequently to modulate the acquisition and development of gut microbiota in early life. The early-life gut microbiota engages in a range of host biological processes, particularly immunity, cognitive neurodevelopment and metabolism. The perturbed early-life gut microbiota increases the risk for disease in early and later life, highlighting the importance of understanding relationships of perinatal factors with early-life microbial composition and functions. In this review, we present an overview of the crucial perinatal factors and summarise updated knowledge of early-life microbiota, as well as how the perinatal factors shape gut microbiota in short and long terms. We further discuss the clinical consequences of perturbations of early-life gut microbiota and potential therapeutic interventions with probiotics/live biotherapeutics.
Collapse
Affiliation(s)
- Shaopu Wang
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| | - Muireann Egan
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| | - C Anthony Ryan
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Department of Paediatrics & Child Health, University College Cork, Cork, Ireland, T12 YN60
| | - Patrick Boyaval
- DuPont Nutrition & Biosciences, Danisco France SAS - DuPont, 22, rue Brunel, F- 75017 Paris, France
| | - Eugene M Dempsey
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Department of Paediatrics & Child Health, University College Cork, Cork, Ireland, T12 YN60
| | - R Paul Ross
- APC Microbiome Ireland, Cork, Ireland, P12 YT20
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland, P12 YT20.,Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland, P61 C996
| |
Collapse
|
182
|
Westaway JAF, Huerlimann R, Miller CM, Kandasamy Y, Norton R, Rudd D. Methods for exploring the faecal microbiome of premature infants: a review. Matern Health Neonatol Perinatol 2021; 7:11. [PMID: 33685524 PMCID: PMC7941982 DOI: 10.1186/s40748-021-00131-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The premature infant gut microbiome plays an important part in infant health and development, and recognition of the implications of microbial dysbiosis in premature infants has prompted significant research into these issues. The approaches to designing investigations into microbial populations are many and varied, each with its own benefits and limitations. The technique used can influence results, contributing to heterogeneity across studies. This review aimed to describe the most common techniques used in researching the preterm infant microbiome, detailing their various limitations. The objective was to provide those entering the field with a broad understanding of available methodologies, so that the likely effects of their use can be factored into literature interpretation and future study design. We found that although many techniques are used for characterising the premature infant microbiome, 16S rRNA short amplicon sequencing is the most common. 16S rRNA short amplicon sequencing has several benefits, including high accuracy, discoverability and high throughput capacity. However, this technique has limitations. Each stage of the protocol offers opportunities for the injection of bias. Bias can contribute to variability between studies using 16S rRNA high throughout sequencing. Thus, we recommend that the interpretation of previous results and future study design be given careful consideration.
Collapse
Affiliation(s)
- Jacob A F Westaway
- James Cook University, 1 McGregor Road, Smithfield, QLD, 4878, Australia.
| | - Roger Huerlimann
- James Cook University, 1 James Cook Dr, Douglas, QLD, 4811, Australia
| | - Catherine M Miller
- James Cook University, 1 McGregor Road, Smithfield, QLD, 4878, Australia
| | - Yoga Kandasamy
- Townsville University Hospital, 100 Angus Smith Dr, Douglas, QLD, 4814, Australia
| | - Robert Norton
- Pathology Queensland, 100 Angus Smith Dr, Douglas, QLD, 4814, Australia
| | - Donna Rudd
- James Cook University, 1 James Cook Dr, Douglas, QLD, 4811, Australia
| |
Collapse
|
183
|
Pang Z, Chen J, Wang T, Gao C, Li Z, Guo L, Xu J, Cheng Y. Linking Plant Secondary Metabolites and Plant Microbiomes: A Review. FRONTIERS IN PLANT SCIENCE 2021; 12:621276. [PMID: 33737943 PMCID: PMC7961088 DOI: 10.3389/fpls.2021.621276] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/08/2021] [Indexed: 05/09/2023]
Abstract
Plant secondary metabolites (PSMs) play many roles including defense against pathogens, pests, and herbivores; response to environmental stresses, and mediating organismal interactions. Similarly, plant microbiomes participate in many of the above-mentioned processes directly or indirectly by regulating plant metabolism. Studies have shown that plants can influence their microbiome by secreting various metabolites and, in turn, the microbiome may also impact the metabolome of the host plant. However, not much is known about the communications between the interacting partners to impact their phenotypic changes. In this article, we review the patterns and potential underlying mechanisms of interactions between PSMs and plant microbiomes. We describe the recent developments in analytical approaches and methods in this field. The applications of these new methods and approaches have increased our understanding of the relationships between PSMs and plant microbiomes. Though the current studies have primarily focused on model organisms, the methods and results obtained so far should help future studies of agriculturally important plants and facilitate the development of methods to manipulate PSMs-microbiome interactions with predictive outcomes for sustainable crop productions.
Collapse
Affiliation(s)
- Zhiqiang Pang
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jia Chen
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Tuhong Wang
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Chunsheng Gao
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Zhimin Li
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Litao Guo
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| | - Jianping Xu
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Yi Cheng
- Institute of Bast Fiber Crops and Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha, China
| |
Collapse
|
184
|
An G, Zhang Y, Fan L, Chen J, Wei M, Li C, Chen X, Zhang L, Yang D, Wang J. Integrative Analysis of Vaginal Microorganisms and Serum Metabolomics in Rats With Estrous Cycle Disorder Induced by Long-Term Heat Exposure Based on 16S rDNA Gene Sequencing and LC/MS-Based Metabolomics. Front Cell Infect Microbiol 2021; 11:595716. [PMID: 33738264 PMCID: PMC7962411 DOI: 10.3389/fcimb.2021.595716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Long term heat exposure (HE) leads to estrous cycle disorder (ECD) in female rats and damages reproductive function. However, the regulation mechanism of vaginal microorganisms and serum metabolomics remains unclear. This study aimed to explore the effects of microbes on the vaginal secretions of rats with ECD and describe the serum metabolomics characteristics and their relationship with vaginal microorganisms. The alterations in the serum levels of neurotransmitters were used to verify the possible regulatory pathways. The relative abundance, composition, and colony interaction network of microorganisms in the vaginal secretions of rats with ECD changed significantly. The metabolomics analysis identified 22 potential biomarkers in the serum including lipid metabolism, amino acid metabolism, and mammalian target of rapamycin and gonadotropin-releasing hormone (GnRH) signaling pathways. Further, 52 pairs of vaginal microbiota–serum metabolites correlations (21 positive and 31 negative) were determined. The abundance of Gardnerella correlated positively with the metabolite L-arginine concentration and negatively with the oleic acid concentration. Further, a negative correlation was found between the abundance of Pseudomonas and the L-arginine concentration and between the metabolite benzoic acid concentration and the abundance of Adlercreutzia. These four bacteria–metabolite pairs had a direct or indirect relationship with the estrous cycle and reproduction. The glutamine, glutamate, and dopamine levels were significantly uncontrolled. The former two were closely related to GnRH signaling pathways involved in the development and regulation of HE-induced ECD in rats. Serum neurotransmitters partly reflected the regulatory effect of vaginal microorganisms on the host of HE-induced ECD, and glutamatergic neurotransmitters might be closely related to the alteration in vaginal microorganisms. These findings might help comprehend the mechanism of HE-induced ECD and propose a new intervention based on vaginal microorganisms.
Collapse
Affiliation(s)
- GaiHong An
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yu Zhang
- Department of Endocrinology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - LiJun Fan
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - JiaJun Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - MengFan Wei
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Chao Li
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - XueWei Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Li Zhang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - DanFeng Yang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Wang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
185
|
Li H, Ma L, Li Z, Yin J, Tan B, Chen J, Jiang Q, Ma X. Evolution of the Gut Microbiota and Its Fermentation Characteristics of Ningxiang Pigs at the Young Stage. Animals (Basel) 2021; 11:ani11030638. [PMID: 33673705 PMCID: PMC7997423 DOI: 10.3390/ani11030638] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/24/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The current study described the evolution of the gut microbiota of an indigenous pig breeds, Ningxiang pigs (NXP), from one week before weaning to the end of nursery. The results showed that dietary factors mainly drove the evolution of the microbial community of NXP. Our results contributed to a better understanding of the evolutionary characteristics and influencing factors of the gut microbiota of indigenous pig breeds. Abstract The current study aimed to investigate the evolution of gut microbiota and its influencing factors for NXP in youth. The results showed that Shannon index increased from d 21 to d 28 whereas the ACE index increased from d 21 until d 60. Firmicutes, mainly Lactobacillus dominated on d 21. The Bacteroides and Spirochetes showed highest relative abundance on d 28. Fiber-degrading bacteria, mainly Prevotellaceae, Lachnospiraceae, Ruminococcaceae, Muribaculaceae, and Oscillospiraceae_UCG−002, dominated the microbial communities at d 28 and d 35. The microbial communities at d 60 and d 75 contained more Clostridium_sensu_stricto_1, Terrisporobacter and Oscillospiraceae_UCG−005 than other ages, which had significantly positive correlations with acetate and total SCFAs concentration. In conclusion, the evolution of gut microbiota was mainly adapted to the change of dietary factors during NXP growth. The response of fiber-degrading bacteria at different stages may help NXP better adapt to plant-derived feeds.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaokang Ma
- Correspondence: ; Tel.: +86-0731-84619706; Fax: +86-0731-84612685
| |
Collapse
|
186
|
Dysbiosis in Pediatrics Is Associated with Respiratory Infections: Is There a Place for Bacterial-Derived Products? Microorganisms 2021; 9:microorganisms9020448. [PMID: 33671586 PMCID: PMC7926342 DOI: 10.3390/microorganisms9020448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory tract infections (RTIs) are common in childhood because of the physiologic immaturity of the immune system, a microbial community under development in addition to other genetic, physiological, environmental and social factors. RTIs tend to recur and severe lower viral RTIs in early childhood are not uncommon and are associated with increased risk of respiratory disorders later in life, including recurrent wheezing and asthma. Therefore, a better understanding of the main players and mechanisms involved in respiratory morbidity is necessary for a prompt and improved care as well as for primary prevention. The inter-talks between human immune components and microbiota as well as their main functions have been recently unraveled; nevertheless, more is still to be discovered or understood in the above medical conditions. The aim of this review paper is to provide the most up-to-date overview on dysbiosis in pre-school children and its association with RTIs and their complications. The potential role of non-harmful bacterial-derived products, according to the old hygiene hypothesis and the most recent trained-innate immunity concept, will be discussed together with the need of proof-of-concept studies and larger clinical trials with immunological and microbiological endpoints.
Collapse
|
187
|
Sarate PJ, Srutkova D, Geissler N, Schwarzer M, Schabussova I, Inic-Kanada A, Kozakova H, Wiedermann U. Pre- and Neonatal Imprinting on Immunological Homeostasis and Epithelial Barrier Integrity by Escherichia coli Nissle 1917 Prevents Allergic Poly-Sensitization in Mice. Front Immunol 2021; 11:612775. [PMID: 33679699 PMCID: PMC7927790 DOI: 10.3389/fimmu.2020.612775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/31/2020] [Indexed: 12/29/2022] Open
Abstract
A steady rise in the number of poly-sensitized patients has increased the demand for effective prophylactic strategies against multi-sensitivities. Probiotic bacteria have been successfully used in clinics and experimental models to prevent allergic mono-sensitization. In the present study, we have investigated whether probiotic bacteria could prevent poly-sensitization by imprinting on the immune system early in life. We used two recombinant variants of probiotic Escherichia coli Nissle 1917 (EcN): i) EcN expressing birch and grass pollen, poly-allergen chimera construct (EcN-Chim), and ii) an “empty” EcN without allergen expression (EcN-Ctrl). Conventional mice (CV) were treated with either EcN-Chim or EcN-Ctrl in the last week of the gestation and lactation period. Gnotobiotic mice received one oral dose of either EcN-Chim or EcN-Ctrl before mating. The offspring from both models underwent systemic allergic poly-sensitization and intranasal challenge with recombinant birch and grass pollen allergens (rBet v 1, rPhl p 1, and rPhl p 5). In the CV setting, the colonization of offspring via treatment of mothers reduced allergic airway inflammation (AAI) in offspring compared to poly-sensitized controls. Similarly, in a gnotobiotic model, AAI was reduced in EcN-Chim and EcN-Ctrl mono-colonized offspring. However, allergy prevention was more pronounced in the EcN-Ctrl mono-colonized offspring as compared to EcN-Chim. Mono-colonization with EcN-Ctrl was associated with a shift toward mixed Th1/Treg immune responses, increased expression of TLR2 and TLR4 in the lung, and maintained levels of zonulin-1 in lung epithelial cells as compared to GF poly-sensitized and EcN-Chim mono-colonized mice. This study is the first one to establish the model of allergic poly-sensitization in gnotobiotic mice. Using two different settings, gnotobiotic and conventional mice, we demonstrated that an early life intervention with the EcN without expressing an allergen is a powerful strategy to prevent poly-sensitization later in life.
Collapse
Affiliation(s)
- Priya J Sarate
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Nora Geissler
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Irma Schabussova
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Aleksandra Inic-Kanada
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hana Kozakova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czechia
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
188
|
Cucinotta U, Romano C, Dipasquale V. Diet and Nutrition in Pediatric Inflammatory Bowel Diseases. Nutrients 2021; 13:655. [PMID: 33671453 PMCID: PMC7922138 DOI: 10.3390/nu13020655] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 12/17/2022] Open
Abstract
Both genetic and environmental factors are involved in the onset of inflammatory bowel disease (IBD). In particular, diet composition is suspected to significantly contribute to IBD risk. In recent years, major interest has raised about the role of nutrition in disease pathogenesis and course, and many studies have shown a clear link between diet composition and intestinal permeability impairment. Moreover, many IBD-related factors, such as poor dietary intake, nutrients loss and drugs interact with nutritional status, thus paving the way for the development of many therapeutic strategies in which nutrition represents the cornerstone, either as first-line therapy or as reversing nutritional deficiencies and malnutrition in IBD patients. Exclusive enteral nutrition (EEN) is the most rigorously supported dietary intervention for the treatment of Crohn's Disease (CD), but is burdened by a low tolerability, especially in pediatric patients. Promising alternative regimens are represented by Crohn's Disease Exclusion Diet (CDED), and other elimination diets, whose use is gradually spreading. The aim of the current paper is to provide a comprehensive and updated overview on the latest evidence about the role of nutrition and diet in pediatric IBD, focusing on the different nutritional interventions available for the management of the disease.
Collapse
Affiliation(s)
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood “G. Barresi”, University of Messina, 98124 Messina, Italy; (U.C.); (V.D.)
| | | |
Collapse
|
189
|
The Multiomics Analyses of Fecal Matrix and Its Significance to Coeliac Disease Gut Profiling. Int J Mol Sci 2021; 22:ijms22041965. [PMID: 33671197 PMCID: PMC7922330 DOI: 10.3390/ijms22041965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal (GIT) diseases have risen globally in recent years, and early detection of the host’s gut microbiota, typically through fecal material, has become a crucial component for rapid diagnosis of such diseases. Human fecal material is a complex substance composed of undigested macromolecules and particles, and the processing of such matter is a challenge due to the unstable nature of its products and the complexity of the matrix. The identification of these products can be used as an indication for present and future diseases; however, many researchers focus on one variable or marker looking for specific biomarkers of disease. Therefore, the combination of genomics, transcriptomics, proteomics and metabonomics can give a detailed and complete insight into the gut environment. The proper sample collection, sample preparation and accurate analytical methods play a crucial role in generating precise microbial data and hypotheses in gut microbiome research, as well as multivariate data analysis in determining the gut microbiome functionality in regard to diseases. This review summarizes fecal sample protocols involved in profiling coeliac disease.
Collapse
|
190
|
Tonon KM, Morais TB, Taddei CR, Araújo-Filho HB, Abrão ACFV, Miranda A, de Morais MB. Gut microbiota comparison of vaginally and cesarean born infants exclusively breastfed by mothers secreting α1-2 fucosylated oligosaccharides in breast milk. PLoS One 2021; 16:e0246839. [PMID: 33556125 PMCID: PMC7870049 DOI: 10.1371/journal.pone.0246839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/26/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exclusive breastfeeding promotes beneficial modifications on the microbiota of cesarean born infants, but little is known about the role of specific breast milk components in this modulation. Women with an active FUT2 gene (called secretors) secrete α1-2 fucosylated human milk oligosaccharides (HMOs), which promote Bifidobacterium in the infant's gut and may modulate the microbiota of cesarean born infants. OBJECTIVE To compare the microbiota composition of cesarean and vaginally born infants breastfed by secretor mothers. METHODS Maternal secretor status was determined by the occurrence of 4 different α1-2 fucosylated HMOs in breast milk by LC-MS. The fecal microbiota composition from cesarean and vaginally born infants was analyzed by 16S rRNA gene sequencing and qPCR, stratified by the maternal secretor status, and compared. RESULTS Alpha and beta diversity were not significantly different in cesarean born, secretor-fed infants (CSe+) compared to vaginally born, secretor-fed infants (VSe+). There were no significant differences in the fecal relative abundance of Bifidobacterium between CSe+ and VSe+ infants, but the prevalence of the species B. longum was lower in CSe+. The fecal relative abundance of Bacteroides was also lower, while Akkermansia and Kluyvera were higher in CSe+ infants. CONCLUSION Cesarean and vaginally born infants fed with breast milk containing the α1-2 fucosylated HMOs fraction present similar amounts of Bifidobacterium in the feces, but differences are observed in other members of the microbiota.
Collapse
Affiliation(s)
- Karina M. Tonon
- Nutrition Postgraduate Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tania B. Morais
- Food Quality Control Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla R. Taddei
- Department of Clinical and Toxicological Analysis, Universidade de São Paulo, São Paulo, Brazil
- School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ana Cristina F. V. Abrão
- Breastfeeding Incentive and Support Center, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Antonio Miranda
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mauro B. de Morais
- Division of Pediatric Gastroenterology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
191
|
Gong C, Yang L, Liu K, Shen S, Zhang Q, Li H, Cheng Y. Effects of Antibiotic Treatment and Probiotics on the Gut Microbiome of 40 Infants Delivered Before Term by Cesarean Section Analysed by Using 16S rRNA Quantitative Polymerase Chain Reaction Sequencing. Med Sci Monit 2021; 27:e928467. [PMID: 33542172 PMCID: PMC7871509 DOI: 10.12659/msm.928467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/19/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effects on the gut microbiome of 40 infants delivered before term by cesarean section between antibiotic treatment and probiotics as assessed by 16S rRNA quantitative polymerase chain reaction (qPCR) sequencing. MATERIAL AND METHODS We divided 40 premature infants delivered by cesarean section into 4 groups according to exposure to antibiotics or probiotics: N group (No-probiotics and No-antibiotics), A group (antibiotics), P group (probiotics), and the AP group (antibiotics+probiotics). Fecal samples were collected on days 1, 3, and 10, and the microflora data were generated using 16S rRNA qPCR sequencing technology. The BugBase tool was used for phenotype prediction, the Tax4Fun tool was used for function prediction, and iPath software was used to predict the metabolic pathways of intestinal bacteria. RESULTS Antibiotics increased the abundance of pathogenic bacteria and reduced the replication and repair function (P=0.049), nucleotide metabolism function (P=0.047), and the purine metabolism pathways (P<0.05) of the gut microbiota. Probiotics increased the abundance of beneficial bacteria and the cellular community prokaryote function (P=0.042) and contributed to the Bifidobacteria biofilm formation. Probiotics alleviated the damage of antibiotics to the composition and function of the gut microbiota. CONCLUSIONS The findings from this study showed that antibiotic treatment of preterm infants born by cesarean section changed the gut microbiome, but that the use of probiotics could restore the normal microbiome, which supports that restoration of the normal gut microbiota may be achieved with probiotics.
Collapse
Affiliation(s)
- Chen Gong
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Kangkang Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Shichun Shen
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Qixing Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Han Li
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
192
|
Mueller NT, Differding MK, Østbye T, Hoyo C, Benjamin-Neelon SE. Association of birth mode of delivery with infant faecal microbiota, potential pathobionts, and short chain fatty acids: a longitudinal study over the first year of life. BJOG 2021; 128:1293-1303. [PMID: 33338292 DOI: 10.1111/1471-0528.16633] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Caesarean section (CS) interrupts mother-to-newborn microbial transfer at birth. Beyond the neonatal period, the impact of CS on offspring gut microbiota and their short-chain fatty acids (SCFAs) remains unclear. Here, we examine birth delivery mode (CS versus vaginal delivery) with the infant gut microbiota and faecal SCFAs measured 3 and 12 months after birth. DESIGN Longitudinal study. SETTING North Carolina. POPULATION In 2013-15, we enrolled pregnant women and followed up their offspring for 12 months. We asked a subset of participants, enrolled over a 3-month period, to provide faecal samples at the 3- and 12-month follow-up visits. METHODS AND MAIN OUTCOMES We sequenced the 16S rRNA V4 region with Illumina MiSeq and quantified SCFA concentrations using gas chromatography. We examined delivery mode with differential abundance of microbiota amplicon sequence variants (ASVs) using beta-binomial regression and faecal SCFAs using linear regression. We adjusted models for confounders. RESULTS Of the 70 infants in our sample, 25 (36%) were delivered by CS. Compared with vaginal delivery, CS was associated with differential abundance of 14 infant bacterial ASVs at 3 months and 13 ASVs at 12 months (all FDR P < 0.05). Of note, CS infants had a higher abundance of the potential pathobionts Clostridium neonatale (P = 0.04) and Clostridium perfringens (P = 0.04) and a lower abundance of potentially beneficial Bifidobacterium and Bacteroides spp. (both P < 0.05) at 3 months. Other ASVs were differentially abundant at 12 months. Infants delivered by CS also had higher faecal butyrate concentration at 3 months (P < 0.005) but not at 12 months. CONCLUSIONS Caesarean section was associated with increased butyrate excretion, decreased Bifidobacterium and Bacteroides spp., and more colonisation of the infant gut by pathobionts at 3 months of age. CS was also associated with altered gut microbiota composition, but not faecal SCFAs, at 12 months. TWEETABLE ABSTRACT Caesarean section delivery was associated with increased butyrate excretion, decreased Bifidobacterium, and increased colonisation of the infant gut by pathobionts at 3 months of age.
Collapse
Affiliation(s)
- N T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - M K Differding
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - T Østbye
- Department of Family Medicine and Community Health, Duke University, Durham, NC, USA
| | - C Hoyo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - S E Benjamin-Neelon
- Department of Health, Behavior and Society, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
193
|
Ni JJ, Yang XL, Zhang H, Xu Q, Wei XT, Feng GJ, Zhao M, Pei YF, Zhang L. Assessing causal relationship from gut microbiota to heel bone mineral density. Bone 2021; 143:115652. [PMID: 32971307 DOI: 10.1016/j.bone.2020.115652] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/28/2022]
Abstract
Recent studies have demonstrated the important role played by gut microbiota in regulating bone development, but the evidence of such causal relationship is still sparse in human population. The aim of this study is to assess the causal relationship from gut microbiota to bone development and to identify specific causal bacteria taxa via a Mendelian randomization (MR) approach. A genome-wide association study (GWAS) summary statistic based two-sample MR analysis was performed. Summary statistics of microbiome GWAS (MGWAS) in 1126 twin pairs of the TwinsUK study was used as discovery sample, and the MGWAS in 984 Dutch participants from the LifeLines-DEEP cohort was used as replication sample. Estimated heel bone mineral density (eBMD) GWAS in 426,824 participants from the UK biobank (UKB) cohort was used as outcome. Bacteria were grouped into taxa features at both order and family levels. In the discovery sample, a total of 25 bacteria features including 9 orders and 16 families were analyzed. Fourteen features (5 orders + 9 families) were nominally significant, including 5 orders (Bacteroidales, Clostridiales, Lactobacillales, Pasteurellales and Verrucomicrobiales) and 9 families (Bacteroidaceae, Clostridiaceae, Lachnospiraceae, Mogibacteriaceae, Pasteurellaceae, Porphyromonadaceae, Streptococcaceae, Verrucomicrobiaceae and Veillonellaceae). One order Clostridiales and its child taxon, family Lachnospiraceae, were successfully replicated in the replication sample (Clostridiales Pdiscovery = 3.32 × 10-3Preplication = 7.29 × 10-3; Lachnospiraceae Pdiscovery = 0.03 Preplication = 7.29 × 10-3). Our findings provided evidence of causal relationship from microbiota to bone development, as well as identified specific bacteria taxa that regulated bone mass variation, thus providing new insights into the microbiota mediated bone development mechanism.
Collapse
Affiliation(s)
- Jing-Jing Ni
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Xiao-Lin Yang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, PR China; Laboratory of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, PR China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Qian Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Xin-Tong Wei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Gui-Juan Feng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Min Zhao
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Yu-Fang Pei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
| |
Collapse
|
194
|
Ratsika A, Codagnone MC, O’Mahony S, Stanton C, Cryan JF. Priming for Life: Early Life Nutrition and the Microbiota-Gut-Brain Axis. Nutrients 2021; 13:423. [PMID: 33525617 PMCID: PMC7912058 DOI: 10.3390/nu13020423] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 12/18/2022] Open
Abstract
Microbes colonize the human body during the first moments of life and coexist with the host throughout the lifespan. Intestinal microbiota and their metabolites aid in the programming of important bodily systems such as the immune and the central nervous system during critical temporal windows of development, with possible structural and functional implications throughout the lifespan. These critical developmental windows perinatally (during the first 1000 days) are susceptible timepoints for insults that can endure long lasting effects on the microbiota-gut-brain axis. Environmental and parental factors like host genetics, mental health, nutrition, delivery and feeding mode, exposure to antibiotics, immune activation and microbiota composition antenatally, are all factors that are able to modulate the microbiota composition of mother and infant and may thus regulate important bodily functions. Among all these factors, early life nutrition plays a pivotal role in perinatal programming and in the modulation of offspring microbiota from birth throughout lifespan. This review aims to present current data on the impact of early life nutrition and microbiota priming of important bodily systems and all the factors influencing the microbial coexistence with the host during early life development.
Collapse
Affiliation(s)
- Anna Ratsika
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland; (A.R.); (M.C.C.); (S.O.); (C.S.)
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 YT20, Ireland
| | - Martin C. Codagnone
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland; (A.R.); (M.C.C.); (S.O.); (C.S.)
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 YT20, Ireland
| | - Siobhain O’Mahony
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland; (A.R.); (M.C.C.); (S.O.); (C.S.)
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 YT20, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland; (A.R.); (M.C.C.); (S.O.); (C.S.)
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12 YT20, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy P61 C996, Ireland
| | - John F. Cryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland; (A.R.); (M.C.C.); (S.O.); (C.S.)
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
195
|
Zhou A, Lei Y, Tang L, Hu S, Yang M, Wu L, Yang S, Tang B. Gut Microbiota: the Emerging Link to Lung Homeostasis and Disease. J Bacteriol 2021; 203:e00454-20. [PMID: 33077630 PMCID: PMC7847545 DOI: 10.1128/jb.00454-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota plays a crucial role in the development of the immune system and confers benefits or disease susceptibility to the host. Emerging studies have indicated the gut microbiota could affect pulmonary health and disease through cross talk between the gut microbiota and the lungs. Gut microbiota dysbiosis could lead to acute or chronic lung disease, such as asthma, tuberculosis, and lung cancer. In addition, the composition of the gut microbiota may be associated with different lung diseases, the prevalence of which also varies by age. Modulation of the gut microbiota through short-chain fatty acids, probiotics, and micronutrients may present potential therapeutic strategies to protect against lung diseases. In this review, we will provide an overview of the cross-talk between the gut microbiota and the lungs, as well as elucidate the underlying pathogenesis and/or potential therapeutic strategies of some lung diseases from the point of view of the gut microbiota.
Collapse
Affiliation(s)
- An Zhou
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuanyuan Lei
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shiping Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Min Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Lingyi Wu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
196
|
Xiong X, Loo SL, Zhang L, Tanaka MM. Modelling the effect of birth and feeding modes on the development of human gut microbiota. Proc Biol Sci 2021; 288:20201810. [PMID: 33434469 PMCID: PMC7892411 DOI: 10.1098/rspb.2020.1810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
The human gut microbiota is transmitted from mother to infant through vaginal birth and breastfeeding. Bifidobacterium, a genus that dominates the infants' gut, is adapted to breast milk in its ability to metabolize human milk oligosaccharides; it is regarded as a mutualist owing to its involvement in the development of the immune system. The composition of microbiota, including the abundance of Bifidobacteria, is highly variable between individuals and some microbial profiles are associated with diseases. However, whether and how birth and feeding practices contribute to such variation remains unclear. To understand how early events affect the establishment of microbiota, we develop a mathematical model of two types of Bifidobacteria and a generic compartment of commensal competitors. We show how early events affect competition between mutualists and commensals and microbe-host-immune interactions to cause long-term alterations in gut microbial profiles. Bifidobacteria associated with breast milk can trigger immune responses with lasting effects on the microbial community structure. Our model shows that, in response to a change in birth environment, competition alone can produce two distinct microbial profiles post-weaning. Adding immune regulation to our competition model allows for variations in microbial profiles in response to different feeding practices. This analysis highlights the importance of microbe-microbe and microbe-host interactions in shaping the gut populations following different birth and feeding modes.
Collapse
Affiliation(s)
- Xiyan Xiong
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | | | | | |
Collapse
|
197
|
Ragonnaud E, Biragyn A. Gut microbiota as the key controllers of "healthy" aging of elderly people. IMMUNITY & AGEING 2021; 18:2. [PMID: 33397404 PMCID: PMC7784378 DOI: 10.1186/s12979-020-00213-w] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
Extrinsic factors, such as lifestyle and diet, are shown to be essential in the control of human healthy aging, and thus, longevity. They do so by targeting at least in part the gut microbiome, a collection of commensal microorganisms (microbiota), which colonize the intestinal tract starting after birth, and is established by the age of three. The composition and abundance of individual microbiota appears to continue to change until adulthood, presumably reflecting lifestyle and geographic, racial, and individual differences. Although most of these changes appear to be harmless, a major shift in their composition in the gut (dysbiosis) can trigger harmful local and systemic inflammation. Recent reports indicate that dysbiosis is increased in aging and that the gut microbiota of elderly people is enriched in pro-inflammatory commensals at the expense of beneficial microbes. The clinical consequence of this change remains confusing due to contradictory reports and a high degree of variability of human microbiota and methodologies used. Here, we present the authors’ thoughts that underscore dysbiosis as a primary cause of aging-associated morbidities, and thus, premature death of elderly people. We provide evidence that the dysbiosis triggers a chain of pathological and inflammatory events. Examples include alteration of levels of microbiota-affected metabolites, impaired function and integrity of the gastrointestinal tract, and increased gut leakiness. All of these enhance systemic inflammation, which when associated with aging is termed inflammaging, and result in consequent aging-associated pathologies.
Collapse
Affiliation(s)
- Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, 251 Bayview Blvd, Suite 100, Baltimore, MD, 21224, USA.
| |
Collapse
|
198
|
Grech A, Collins CE, Holmes A, Lal R, Duncanson K, Taylor R, Gordon A. Maternal exposures and the infant gut microbiome: a systematic review with meta-analysis. Gut Microbes 2021; 13:1-30. [PMID: 33978558 PMCID: PMC8276657 DOI: 10.1080/19490976.2021.1897210] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 02/04/2023] Open
Abstract
Early life, including the establishment of the intestinal microbiome, represents a critical window of growth and development. Postnatal factors affecting the microbiome, including mode of delivery, feeding type, and antibiotic exposure have been widely investigated, but questions remain regarding the influence of exposures in utero on infant gut microbiome assembly. This systematic review aimed to synthesize evidence on exposures before birth, which affect the early intestinal microbiome. Five databases were searched in August 2019 for studies exploring pre-pregnancy or pregnancy 'exposure' data in relation to the infant microbiome. Of 1,441 publications identified, 76 were included. Factors reported influencing microbiome composition and diversity included maternal antibiotic and probiotic uses, dietary intake, pre-pregnancy body mass index (BMI), gestational weight gain (GWG), diabetes, mood, and others. Eleven studies contributed to three meta-analyses quantifying associations between maternal intrapartum antibiotic exposure (IAP), BMI and GWG, and infant microbiome alpha diversity (Shannon Index). IAP, maternal overweight/obesity and excessive GWG were all associated with reduced diversity. Most studies were observational, few included early recruitment or longitudinal follow-up, and the timing, frequency, and methodologies related to stool sampling and analysis were variable. Standardization and collaboration are imperative to enhance understanding in this complex and rapidly evolving area.
Collapse
Affiliation(s)
- Allison Grech
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Clare E Collins
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW, Australia
| | - Andrew Holmes
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Camperdown, NSW, Australia
| | - Ravin Lal
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Kerith Duncanson
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Rachael Taylor
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW, Australia
| | - Adrienne Gordon
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales(NSW), Australia
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
199
|
Egan M, Dempsey E, Ryan CA, Ross RP, Stanton C. The Sporobiota of the Human Gut. Gut Microbes 2021; 13:1-17. [PMID: 33406976 PMCID: PMC7801112 DOI: 10.1080/19490976.2020.1863134] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/04/2023] Open
Abstract
The human gut microbiome is a diverse and complex ecosystem that plays a critical role in health and disease. The composition of the gut microbiome has been well studied across all stages of life. In recent years, studies have investigated the production of endospores by specific members of the gut microbiome. An endospore is a tough, dormant structure formed by members of the Firmicutes phylum, which allows for greater resistance to otherwise inhospitable conditions. This innate resistance has consequences for human health and disease, as well as in biotechnology. In particular, the formation of endospores is strongly linked to antibiotic resistance and the spread of antibiotic resistance genes, also known as the resistome. The term sporobiota has been used to define the spore-forming cohort of a microbial community. In this review, we present an overview of the current knowledge of the sporobiota in the human gut. We discuss the development of the sporobiota in the infant gut and the perinatal factors that may have an effect on vertical transmission from mother to infant. Finally, we examine the sporobiota of critically important food sources for the developing infant, breast milk and powdered infant formula.
Collapse
Affiliation(s)
- Muireann Egan
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
200
|
Wang X, Li J, Li N, Guan K, Yin D, Zhang H, Ding G, Hu Y. Evolution of Intestinal Gases and Fecal Short-Chain Fatty Acids Produced in vitro by Preterm Infant Gut Microbiota During the First 4 Weeks of Life. Front Pediatr 2021; 9:726193. [PMID: 34646797 PMCID: PMC8504453 DOI: 10.3389/fped.2021.726193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/27/2021] [Indexed: 12/31/2022] Open
Abstract
Background: The production of intestinal gases and fecal short-chain fatty acids (SCFAs) by infant gut microbiota may have a significant impact on their health, but information about the composition and volume of intestinal gases and SCFA profiles in preterm infants is scarce. Objective: This study examined the change of the composition and volume of intestinal gases and SCFA profiles produced by preterm infant gut microbiota in vitro during the first 4 weeks of life. Methods: Fecal samples were obtained at five time points (within 3 days, 1 week, 2 weeks, 3 weeks, and 4 weeks) from 19 preterm infants hospitalized in the neonatal intensive care unit (NICU) of Shanghai Children's Hospital, Shanghai Jiao Tong University between May and July 2020. These samples were initially inoculated into four different media containing lactose (LAT), fructooligosaccharide (FOS), 2'-fucosyllactose (FL-2), and galactooligosaccharide (GOS) and thereafter fermented for 24 h under conditions mimicking those of the large intestine at 37.8°C under anaerobic conditions. The volume of total intestinal gases and the concentrations of individual carbon dioxide (CO2), hydrogen (H2), methane (CH4), and hydrogen sulfide (H2S) were measured by a gas analyzer. The concentrations of total SCFAs, individual acetic acid, propanoic acid, butyric acid, isobutyric acid, pentanoic acid, and valeric acid were measured by gas chromatography (GC). Results: The total volume of intestinal gases (ranging from 0.01 to 1.64 ml in medium with LAT; 0-1.42 ml with GOS; 0-0.91 ml with FOS; and 0-0.44 ml with FL-2) and the concentrations of CO2, H2, H2S, and all six fecal SCFAs increased with age (p-trends < 0.05). Among them, CO2 was usually the predominant intestinal gas, and acetic acid was usually the predominant SCFA. When stratified by birth weight (<1,500 and ≥1,500 g), gender, and delivery mode, the concentration of CO2 was more pronounced among infants whose weight was ≥1,500 g than among those whose weight was <1,500 g (p-trends < 0.05). Conclusions: Our findings suggested that the intestinal gases and SCFAs produced by preterm infant gut microbiota in vitro increased with age during the first 4 weeks of life.
Collapse
Affiliation(s)
- Xuefang Wang
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Juan Li
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Na Li
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kunyu Guan
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Di Yin
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huating Zhang
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guodong Ding
- Department of Respiratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Hu
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|