201
|
Girma A. The effect of novel antimicrobial agents on the normal functioning of human intestinal microbiota: a systematic review. FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1159352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Antimicrobial agents have significant effects on the ecological balance of the human microbiota through incomplete absorption (e.g., orally administered antimicrobial agents) or secretion (e.g., by the salivary glands, in the bile, or from the intestinal mucosa) of the agents. This study aimed to examine the effects of novel antimicrobial agents on the normal functioning of the intestinal microbiota. The articles, written in English, were recovered from PubMed, ScienceDirect, Web of Science, Google Scholar, and DOAJ, as well as from manual searches using a reference list. “Microbiota”, “Intestinal Microbiota”, “Eubiotic Microbiota”, “Ecological Impact”, “Antimicrobial Agents,”, “Antibiotics”, “Dysbiosis”, “Gut Microbiota”, and “Probiotics” were the search terms used to retrieve the articles. The PRISMA 2009 checklist was applied for article search strategy, article selection, data extraction, and result reporting for the review process. A total of eight original research articles were included from a total of 379 articles obtained in different search strategies. The eight new antimicrobial agents demonstrated significant impacts on the ecological balance of the human intestinal microbiota. Therefore, eubiosis is crucial in preventing the establishment of exogenous antimicrobial-resistant strains as well as their gene transfer.Systematic review registration[PRISMA], identifier [2009].
Collapse
|
202
|
Querdasi FR, Vogel SC, Thomason ME, Callaghan BL, Brito NH. A comparison of the infant gut microbiome before versus after the start of the covid-19 pandemic. Sci Rep 2023; 13:13289. [PMID: 37587195 PMCID: PMC10432475 DOI: 10.1038/s41598-023-40102-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
The COVID-19 pandemic and resulting public health directives led to many changes in families' social and material environments. Prior research suggests that these changes are likely to impact composition of the gut microbiome, particularly during early childhood when the gut microbiome is developing most rapidly. Importantly, disruption to the gut microbiome during this sensitive period can have potentially long-lasting impacts on health and development. In the current study, we compare gut microbiome composition among a socioeconomically and racially diverse group of 12-month old infants living in New York City who provided stool samples before the pandemic (N = 34) to a group who provided samples during the first 9-months of the pandemic (March-December 2020; N = 20). We found that infants sampled during the pandemic had lower alpha diversity of the microbiome, lower abundance of Pasteurellaceae and Haemophilus, and significantly different beta diversity based on unweighted Unifrac distance than infants sampled before the pandemic. Exploratory analyses suggest that gut microbiome changes due to the pandemic occurred relatively quickly after the start of the pandemic and were sustained. Our results provide evidence that pandemic-related environmental disruptions had an impact on community-level taxonomic diversity of the developing gut microbiome, as well as abundance of specific members of the gut bacterial community.
Collapse
|
203
|
Mangiola S, Roth-Schulze AJ, Trussart M, Zozaya-Valdés E, Ma M, Gao Z, Rubin AF, Speed TP, Shim H, Papenfuss AT. sccomp: Robust differential composition and variability analysis for single-cell data. Proc Natl Acad Sci U S A 2023; 120:e2203828120. [PMID: 37549298 PMCID: PMC10438834 DOI: 10.1073/pnas.2203828120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/18/2023] [Indexed: 08/09/2023] Open
Abstract
Cellular omics such as single-cell genomics, proteomics, and microbiomics allow the characterization of tissue and microbial community composition, which can be compared between conditions to identify biological drivers. This strategy has been critical to revealing markers of disease progression, such as cancer and pathogen infection. A dedicated statistical method for differential variability analysis is lacking for cellular omics data, and existing methods for differential composition analysis do not model some compositional data properties, suggesting there is room to improve model performance. Here, we introduce sccomp, a method for differential composition and variability analyses that jointly models data count distribution, compositionality, group-specific variability, and proportion mean-variability association, being aware of outliers. sccomp provides a comprehensive analysis framework that offers realistic data simulation and cross-study knowledge transfer. Here, we demonstrate that mean-variability association is ubiquitous across technologies, highlighting the inadequacy of the very popular Dirichlet-multinomial distribution. We show that sccomp accurately fits experimental data, significantly improving performance over state-of-the-art algorithms. Using sccomp, we identified differential constraints and composition in the microenvironment of primary breast cancer.
Collapse
Affiliation(s)
- Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC3052, Australia
| | - Alexandra J. Roth-Schulze
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC3052, Australia
| | - Marie Trussart
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
| | - Enrique Zozaya-Valdés
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC3052, Australia
| | - Mengyao Ma
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
| | - Zijie Gao
- Melbourne Integrative Genomics, University of Melbourne, Parkville, VIC3052, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC3052, Australia
| | - Alan F. Rubin
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC3052, Australia
| | - Terence P. Speed
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
| | - Heejung Shim
- Melbourne Integrative Genomics, University of Melbourne, Parkville, VIC3052, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC3052, Australia
| | - Anthony T. Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC3052, Australia
| |
Collapse
|
204
|
Buzun E, Hsu CY, Sejane K, Oles RE, Ayala AV, Loomis LR, Zhao J, Rossitto LA, McGrosso D, Gonzalez DJ, Bode L, Chu H. A bacterial sialidase mediates early life colonization by a pioneering gut commensal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552477. [PMID: 37609270 PMCID: PMC10441351 DOI: 10.1101/2023.08.08.552477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The early microbial colonization of the gastrointestinal tract can lead to long-term impacts in development and overall human health. Keystone species, including Bacteroides spp ., play a crucial role in maintaining the structure, diversity, and function of the intestinal ecosystem. However, the process by which a defined and resilient community is curated and maintained during early life remains inadequately understood. Here, we show that a single sialidase, NanH, in Bacteroides fragilis mediates stable occupancy of the intestinal mucosa and regulates the commensal colonization program during the first weeks of life. This program is triggered by sialylated glycans, including those found in human milk oligosaccharides and intestinal mucus. After examining the dynamics between pioneer gut Bacteroides species in the murine gut, we discovered that NanH enables vertical transmission from dams to pups and promotes B. fragilis dominance during early life. Furthermore, we demonstrate that NanH facilitates commensal resilience and recovery after antibiotic treatment in a defined microbial community. Collectively, our study reveals a co-evolutionary mechanism between the host and the microbiota mediated through host-derived glycans to promote stable intestinal colonization.
Collapse
|
205
|
Beghetti I, Barone M, Brigidi P, Sansavini A, Corvaglia L, Aceti A, Turroni S. Early-life gut microbiota and neurodevelopment in preterm infants: a narrative review. Front Nutr 2023; 10:1241303. [PMID: 37614746 PMCID: PMC10443645 DOI: 10.3389/fnut.2023.1241303] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/27/2023] [Indexed: 08/25/2023] Open
Abstract
Infants born preterm are at a high risk of both gut microbiota (GM) dysbiosis and neurodevelopmental impairment. While the link between early dysbiosis and short-term clinical outcomes is well established, the relationship with long-term infant health has only recently gained interest. Notably, there is a significant overlap in the developmental windows of GM and the nervous system in early life. The connection between GM and neurodevelopment was first described in animal models, but over the last decade a growing body of research has also identified GM features as one of the potential mediators for human neurodevelopmental and neuropsychiatric disorders. In this narrative review, we provide an overview of the developing GM in early life and its prospective relationship with neurodevelopment, with a focus on preterm infants. Animal models have provided evidence for emerging pathways linking early-life GM with brain development. Furthermore, a relationship between both dynamic patterns and static features of the GM during preterm infants' early life and brain maturation, as well as neurodevelopmental outcomes in early childhood, was documented. Future human studies in larger cohorts, integrated with studies on animal models, may provide additional evidence and help to identify predictive biomarkers and potential therapeutic targets for healthy neurodevelopment in preterm infants.
Collapse
Affiliation(s)
- Isadora Beghetti
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Monica Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Sansavini
- Department of Psychology “Renzo Canestrari”, University of Bologna, Bologna, Italy
| | - Luigi Corvaglia
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Arianna Aceti
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
206
|
Luzardo-Ocampo I, Ocampo-Ruiz AL, Dena-Beltrán JL, Martínez de la Escalera G, Clapp C, Macotela Y. The Diversity of Gut Microbiota at Weaning Is Altered in Prolactin Receptor-Null Mice. Nutrients 2023; 15:3447. [PMID: 37571383 PMCID: PMC10420910 DOI: 10.3390/nu15153447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Maternal milk supports offspring development by providing microbiota, macronutrients, micronutrients, immune factors, and hormones. The hormone prolactin (PRL) is an important milk component with protective effects against metabolic diseases. Because maternal milk regulates microbiota composition and adequate microbiota protect against the development of metabolic diseases, we aimed to investigate whether PRL/PRL receptor signaling regulates gut microbiota composition in newborn mice at weaning. 16SrRNA sequencing of feces and bioinformatics analysis was performed to evaluate gut microbiota in PRL receptor-null mice (Prlr-KO) at weaning (postnatal day 21). The normalized colon and cecal weights were higher and lower, respectively, in the Prlr-KO mice relative to the wild-type mice (Prlr-WT). Relative abundances (Simpson Evenness Index), phylogenetic diversity, and bacterial concentrations were lower in the Prlr-KO mice. Eleven bacteria species out of 470 differed between the Prlr-KO and Prlr-WT mice, with two genera (Anaerotruncus and Lachnospiraceae) related to metabolic disease development being the most common in the Prlr-KO mice. A higher metabolism of terpenoids and polyketides was predicted in the Prlr-KO mice compared to the Prlr-WT mice, and these metabolites had antimicrobial properties and were present in microbe-associated pathogenicity. We concluded that the absence of the PRL receptor altered gut microbiota, resulting in lower abundance and richness, which could contribute to metabolic disease development.
Collapse
Affiliation(s)
| | | | | | | | | | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro 76230, Mexico; (I.L.-O.); (A.L.O.-R.); (J.L.D.-B.); (G.M.d.l.E.); (C.C.)
| |
Collapse
|
207
|
Hoel ST, Wiik J, Carlsen KC, Endre KM, Gudmundsdóttir HK, Haugen G, Hoyer A, Jonassen CM, LeBlanc M, Nordlund B, Rudi K, Skjerven HO, Staff AC, Hedlin G, Söderhäll C, Vettukattil R, Aaneland H, Rehbinder EM. Birth mode is associated with development of atopic dermatitis in infancy and early childhood. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100104. [PMID: 37779526 PMCID: PMC10509990 DOI: 10.1016/j.jacig.2023.100104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/15/2023] [Accepted: 02/05/2023] [Indexed: 10/03/2023]
Abstract
Background Birth by caesarean section (CS) is associated with development of allergic diseases, but its role in the development of atopic dermatitis (AD) is less convincing. Objective Our primary aim was to determine if birth mode was associated with AD in 3-year-olds and secondarily to determine if birth mode was associated with early onset and/or persistent AD in the first 3 years of life. Methods We included 2129 mother-child pairs from the Scandinavian population-based prospective PreventADALL cohort with information on birth mode including vaginal birth, either traditional (81.3%) or in water (4.0%), and CS before (6.3%) and after (8.5%) onset of labor. We defined early onset AD as eczema at 3 months and AD diagnosis by 3 years of age. Persistent AD was defined as eczema both in the first year and at 3 years of age, together with an AD diagnosis by 3 years of age. Results AD was diagnosed at 3, 6, 12, 24, and/or 36 months in 531 children (25%). Compared to vaginal delivery, CS was overall associated with increased odds of AD by 3 years of age, with adjusted odds ratio (95% confidence interval) of 1.33 (1.02-1.74), and higher odds of early onset AD (1.63, 1.06-2.48). The highest odds for early onset AD were observed in infants born by CS after onset of labor (1.83, 1.09-3.07). Birth mode was not associated with persistent AD. Conclusion CS was associated with increased odds of AD by 3 years of age, particularly in infants presenting with eczema at 3 months of age.
Collapse
Affiliation(s)
- Sveinung T. Hoel
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Johanna Wiik
- Department of Gynecology and Obstetrics, Østfold Hospital Trust, Kalnes, Norway
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Karin C.L. Carlsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Kim M.A. Endre
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Dermatology and Vaenerology, Oslo University Hospital, Oslo, Norway
| | - Hrefna Katrín Gudmundsdóttir
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Guttorm Haugen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Angela Hoyer
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| | - Christine Monceyron Jonassen
- Centre for Laboratory Medicine, Østfold Hospital Trust, Grålum, Norway
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Marissa LeBlanc
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Björn Nordlund
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Håvard O. Skjerven
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Anne Cathrine Staff
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Gunilla Hedlin
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| | - Cilla Söderhäll
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institute, Stockholm, Sweden
| | - Riyas Vettukattil
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Hilde Aaneland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Eva M. Rehbinder
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Dermatology and Vaenerology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
208
|
Jokela R, Ponsero AJ, Dikareva E, Wei X, Kolho KL, Korpela K, de Vos WM, Salonen A. Sources of gut microbiota variation in a large longitudinal Finnish infant cohort. EBioMedicine 2023; 94:104695. [PMID: 37399600 PMCID: PMC10328818 DOI: 10.1016/j.ebiom.2023.104695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Although the infant gut microbiota has been extensively studied, comprehensive assessment on the microbiota determinants including technical variables has not been performed in large infant cohorts. METHODS We studied the effect of 109 variables on the 16S rRNA gene amplicon-based gut microbiota profiles of infants sampled longitudinally from three weeks to two years of life in the Finnish HELMi birth cohort. Spot faecal samples from both parents were included for intra-family analyses, totalling to 7657 samples from 985 families that were evaluated for beta-diversity patterns using permutational multivariate analysis on Bray-Curtis distances, and differential abundance testing and alpha-diversity for variables of interest. We also assessed the effect of different taxonomic levels and distance methods. FINDINGS In time point-specific models, the largest share of variation explained, up to 2-6%, were seen in decreasing order for the DNA extraction batch, delivery mode and related perinatal exposures, defecation frequency and parity/siblings. Variables describing the infant gastrointestinal function were continuously important during the first two years, reflecting changes in e.g., feeding habits. The effect of parity/siblings on infant microbiota was modified by birth mode and exposure to intrapartum antibiotics, exemplifying the tight interlinkage of perinatal factors relevant for infant microbiota research. In total, up to 19% of the biological microbiota variation in the infant gut could be explained. Our results highlight the need to interpret variance partitioning results in the context of each cohort's characteristics and microbiota processing. INTERPRETATION Our study provides a comprehensive report of key factors associated with infant gut microbiota composition across the two first years of life in a homogenous cohort. The study highlights possible important future research areas and confounding factors to be considered. FUNDING This research was supported by Business Finland, Academy of Finland, Foundation for Nutrition Research and the Doctoral Program in Microbiology and Biotechnology, University of Helsinki, Finland.
Collapse
Affiliation(s)
- Roosa Jokela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alise J Ponsero
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Evgenia Dikareva
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Xiaodong Wei
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kaija-Leena Kolho
- Children's Hospital, Paediatric Research Centre, University of Helsinki and HUS, Helsinki, Finland; Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
209
|
Peppas I, Ford AM, Furness CL, Greaves MF. Gut microbiome immaturity and childhood acute lymphoblastic leukaemia. Nat Rev Cancer 2023; 23:565-576. [PMID: 37280427 PMCID: PMC10243253 DOI: 10.1038/s41568-023-00584-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2023] [Indexed: 06/08/2023]
Abstract
Acute lymphoblastic leukaemia (ALL) is the most common cancer of childhood. Here, we map emerging evidence suggesting that children with ALL at the time of diagnosis may have a delayed maturation of the gut microbiome compared with healthy children. This finding may be associated with early-life epidemiological factors previously identified as risk indicators for childhood ALL, including caesarean section birth, diminished breast feeding and paucity of social contacts. The consistently observed deficiency in short-chain fatty-acid-producing bacterial taxa in children with ALL has the potential to promote dysregulated immune responses and to, ultimately, increase the risk of transformation of preleukaemic clones in response to common infectious triggers. These data endorse the concept that a microbiome deficit in early life may contribute to the development of the major subtypes of childhood ALL and encourage the notion of risk-reducing microbiome-targeted intervention in the future.
Collapse
Affiliation(s)
- Ioannis Peppas
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Department of Paediatric Oncology, The Royal Marsden Hospital Sutton, Surrey, UK
| | - Anthony M Ford
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Caroline L Furness
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Department of Paediatric Oncology, The Royal Marsden Hospital Sutton, Surrey, UK
| | - Mel F Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
210
|
Huang H, Zhong W, Wang X, Yang Y, Wu T, Chen R, Liu Y, He F, Li J. The role of gastric microecological dysbiosis in gastric carcinogenesis. Front Microbiol 2023; 14:1218395. [PMID: 37583514 PMCID: PMC10423824 DOI: 10.3389/fmicb.2023.1218395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Gastric cancer (GC) is the leading cause of cancer-related death worldwide, and reducing its mortality has become an urgent public health issue. Gastric microecological dysbiosis (including bacteria, fungi, viruses, acid suppressants, antibiotics, and surgery) can lead to gastric immune dysfunction or result in a decrease in dominant bacteria and an increase in the number and virulence of pathogenic microorganisms, which in turn promotes development of GC. This review analyzes the relationship between gastric microecological dysbiosis and GC, elucidates dynamic alterations of the microbiota in Correa's cascade, and identifies certain specific microorganisms as potential biomarkers of GC to aid in early screening and diagnosis. In addition, this paper presents the potential of gastric microbiota transplantation as a therapeutic target for gastric cancer, providing a new direction for future research in this field.
Collapse
Affiliation(s)
- Hui Huang
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Wei Zhong
- Chengdu Medical College, Chengdu, Sichuan, China
| | | | - Ying Yang
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Tianmu Wu
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Runyang Chen
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Yanling Liu
- Chengdu Medical College, Chengdu, Sichuan, China
| | - Feng He
- Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Jun Li
- Chengdu Medical College, Chengdu, Sichuan, China
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
211
|
Saturio S, Rey A, Samarra A, Collado MC, Suárez M, Mantecón L, Solís G, Gueimonde M, Arboleya S. Old Folks, Bad Boon: Antimicrobial Resistance in the Infant Gut Microbiome. Microorganisms 2023; 11:1907. [PMID: 37630467 PMCID: PMC10458625 DOI: 10.3390/microorganisms11081907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
The development of the intestinal microbiome in the neonate starts, mainly, at birth, when the infant receives its founding microbial inoculum from the mother. This microbiome contains genes conferring resistance to antibiotics since these are found in some of the microorganisms present in the intestine. Similarly to microbiota composition, the possession of antibiotic resistance genes is affected by different perinatal factors. Moreover, antibiotics are the most used drugs in early life, and the use of antibiotics in pediatrics covers a wide variety of possibilities and treatment options. The disruption in the early microbiota caused by antibiotics may be of great relevance, not just because it may limit colonization by beneficial microorganisms and increase that of potential pathogens, but also because it may increase the levels of antibiotic resistance genes. The increase in antibiotic-resistant microorganisms is one of the major public health threats that humanity has to face and, therefore, understanding the factors that determine the development of the resistome in early life is of relevance. Recent advancements in sequencing technologies have enabled the study of the microbiota and the resistome at unprecedent levels. These aspects are discussed in this review as well as some potential interventions aimed at reducing the possession of resistance genes.
Collapse
Affiliation(s)
- Silvia Saturio
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| | - Alejandra Rey
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
| | - Anna Samarra
- Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980 Paterna, Spain; (A.S.); (M.C.C.)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980 Paterna, Spain; (A.S.); (M.C.C.)
| | - Marta Suárez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Laura Mantecón
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Gonzalo Solís
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| | - Silvia Arboleya
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| |
Collapse
|
212
|
Fu G, Ma G, Dou S, Wang Q, Fu L, Zhang X, Lu C, Cong B, Li S. Feature selection with a genetic algorithm can help improve the distinguishing power of microbiota information in monozygotic twins' identification. Front Microbiol 2023; 14:1210638. [PMID: 37555059 PMCID: PMC10406218 DOI: 10.3389/fmicb.2023.1210638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
INTRODUCTION Personal identification of monozygotic twins (MZT) has been challenging in forensic genetics. Previous research has demonstrated that microbial markers have potential value due to their specificity and long-term stability. However, those studies would use the complete information of detected microbial communities, and low-value species would limit the performance of previous models. METHODS To address this issue, we collected 80 saliva samples from 10 pairs of MZTs at four different time points and used 16s rRNA V3-V4 region sequencing to obtain microbiota information. The data formed 280 inner-individual (Self) or MZT sample pairs, divided into four groups based on the individual relationship and time interval, and then randomly divided into training and testing sets with an 8:2 ratio. We built 12 identification models based on the time interval ( ≤ 1 year or ≥ 2 months), data basis (Amplicon sequence variants, ASVs or Operational taxonomic unit, OTUs), and distance parameter selection (Jaccard distance, Bray-Curist distance, or Hellinger distance) and then improved their identification power through genetic algorithm processes. The best combination of databases with distance parameters was selected as the final model for the two types of time intervals. Bayes theory was introduced to provide a numerical indicator of the evidence's effectiveness in practical cases. RESULTS From the 80 saliva samples, 369 OTUs and 1130 ASVs were detected. After the feature selection process, ASV-Jaccard distance models were selected as the final models for the two types of time intervals. For short interval samples, the final model can completely distinguish MZT pairs from Self ones in both training and test sets. DISCUSSION Our findings support the microbiota solution to the challenging MZT identification problem and highlight the importance of feature selection in improving model performance.
Collapse
Affiliation(s)
- Guangping Fu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Guanju Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Shujie Dou
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Qian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Lihong Fu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Xiaojing Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Chaolong Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- Hainan Tropical Forensic Medicine Academician Workstation, Haikou, China
| | - Shujin Li
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| |
Collapse
|
213
|
Moy M, Diakiw L, Amato KR. Human-influenced diets affect the gut microbiome of wild baboons. Sci Rep 2023; 13:11886. [PMID: 37482555 PMCID: PMC10363530 DOI: 10.1038/s41598-023-38895-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 07/17/2023] [Indexed: 07/25/2023] Open
Abstract
Industrialized diets that incorporate processed foods and are often high in simple sugars and fats and low in fiber have myriad health impacts, many of which may operate via impacts on the gut microbiota. Examining how these diets affect the gut microbiota can be challenging given that lab animal models experience altered environmental contexts, and human studies include a suite of co-varying cultural and environmental factors that are likely to shape the gut microbiota alongside diet. To complement these approaches, we compare the microbiomes of wild populations of olive baboons (Papio anubis) with differential access to human trash high in processed foods, simple sugars, and fats in Rwanda's Akagera National Park. Baboons are a good model system since their microbiomes are compositionally similar to those of humans. Additionally, this population inhabits a common environment with different social groups consuming qualitatively different amounts of human trash, limiting variation in non-dietary factors. Using 16S rRNA gene amplicon sequencing we find that baboons with unlimited access to human trash have reduced microbial alpha diversity and reduced relative abundances of fiber-degrading taxa such as Ruminococcaceae, Prevotellaceae, and Lachnospiraceae. In contrast, baboons with limited access to human trash have a microbiome more similar to that of baboons with no access to human trash. Our results suggest that while a human-influenced diet high in processed foods, simple sugars, and fats is sufficient to alter the microbiome in wild baboons, there is a minimum threshold of dietary alteration that must occur before the microbiome is substantially altered. We recommend that data from wild primate populations such as these be used to complement ongoing research on diet-microbiome-health interactions in humans and lab animal models.
Collapse
Affiliation(s)
- Madelyn Moy
- Department of Anthropology, Northwestern University, Evanston, IL, 60208, USA
| | - Laura Diakiw
- Department of Ecology, University of Wyoming, Laramie, WY, 82071, USA
| | - Katherine R Amato
- Department of Anthropology, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
214
|
Chang YS, Li CW, Chen L, Wang XA, Lee MS, Chao YH. Early Gut Microbiota Profile in Healthy Neonates: Microbiome Analysis of the First-Pass Meconium Using Next-Generation Sequencing Technology. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1260. [PMID: 37508757 PMCID: PMC10377966 DOI: 10.3390/children10071260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Gut microbiome development during early life has significant long-term effects on health later in life. The first-pass meconium is not sterile, and it is important to know the initial founder of the subsequent gut microbiome. However, there is limited data on the microbiota profile of the first-pass meconium in healthy neonates. To determine the early gut microbiota profile, we analyzed 39 samples of the first-pass meconium from healthy neonates using 16S rRNA sequencing. Our results showed a similar profile of the microbiota composition in the first-pass meconium samples. Pseudomonas was the most abundant genus in most samples. The evenness of the microbial communities in the first-pass meconium was extremely poor, and the average Shannon diversity index was 1.31. An analysis of the relationship between perinatal characteristics and the meconium microbiome revealed that primigravidae babies had a significantly higher Shannon diversity index (p = 0.041), and the Bacteroidales order was a biomarker for the first-pass meconium of these neonates. The Shannon diversity index was not affected by the mode of delivery, maternal intrapartum antibiotic treatment, prolonged rupture of membranes, or birth weight. Our study extends previous research with further characterization of the gut microbiome in very early life.
Collapse
Affiliation(s)
- Yi-Sheng Chang
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Chang-Wei Li
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Ling Chen
- Department of Research and Development, AllBio Life Incorporation, Taichung 402, Taiwan
| | - Xing-An Wang
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Maw-Sheng Lee
- Department of Obstetrics and Gynecology, Lee Women's Hospital, Taichung 406, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Clinical Pathology, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| |
Collapse
|
215
|
Dovrolis N, Moschoviti A, Fessatou S, Karamanolis G, Kolios G, Gazouli M. Identifying Microbiome Dynamics in Pediatric IBD: More than a Family Matter. Biomedicines 2023; 11:1979. [PMID: 37509618 PMCID: PMC10377534 DOI: 10.3390/biomedicines11071979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Pediatric inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease that affects both children and adolescents. Symptoms can significantly affect a child's growth, development, and quality of life, making early diagnosis and effective management crucial. This study focuses on treatment-naïve pediatric IBD patients and their immediate families to identify the role of the microbiome in disease onset. METHODS Nine families with pediatric IBD were recruited, comprising seven drug-naïve Crohn's disease (CD) patients and two drug-naïve ulcerative colitis (UC) patients, as well as twenty-four healthy siblings/parents. Fecal samples were collected for 16S ribosomal RNA gene sequencing and bioinformatics analysis. RESULTS We identified patterns of dysbiosis and hallmark microbial taxa among patients who shared ethnic, habitual, and dietary traits with themselves and their families. In addition, we examined the impact of the disease on specific microbial taxa and how these could serve as potential biomarkers for early detection. CONCLUSIONS Our results suggest a potential role of maternal factors in the establishment and modulation of the early life microbiome, consistent with the current literature, which may have implications for understanding the etiology and progression of IBD.
Collapse
Affiliation(s)
- Nikolas Dovrolis
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.D.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Anastasia Moschoviti
- Third Department of Pediatrics, “Attikon” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.M.); (S.F.)
| | - Smaragdi Fessatou
- Third Department of Pediatrics, “Attikon” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.M.); (S.F.)
| | - George Karamanolis
- Gastroenterology Unit, Second Department of Surgery, Aretaieio Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.D.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- School of Science and Technology, Hellenic Open University, 26335 Patra, Greece
| |
Collapse
|
216
|
Jaspan HB, Mitchell CM, Happel AU. The vagina question: Can maternal vaginal fluid impact the infant gut microbiome and neurodevelopment? Cell Host Microbe 2023; 31:1084-1086. [PMID: 37442096 DOI: 10.1016/j.chom.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cesarean section rates continue to rise globally, and C-sectioned infants are at a higher risk of adverse child outcomes. In this issue of Cell Host & Microbe, Zhou et al. report that vaginal microbial transfer (VMT) from birth mother to infant post-delivery may alter infant gut microbiota and improve neurodevelopment.
Collapse
Affiliation(s)
- Heather B Jaspan
- Center for Global Infectious Diseases Research, Seattle Children's Research Institute, Seattle, WA, USA; Departments of Pediatrics and Global Health, University of Washington, Seattle, WA, USA; Department of Pathology, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| | - Caroline M Mitchell
- Department of Obstetrics & Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Anna-Ursula Happel
- Department of Pathology, University of Cape Town, Cape Town, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
217
|
Liu Y, Ma J, Zhu B, Liu F, Qin S, Lv N, Feng Y, Wang S, Yang H. A health-promoting role of exclusive breastfeeding on infants through restoring delivery mode-induced gut microbiota perturbations. Front Microbiol 2023; 14:1163269. [PMID: 37492252 PMCID: PMC10363731 DOI: 10.3389/fmicb.2023.1163269] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/15/2023] [Indexed: 07/27/2023] Open
Abstract
The establishment of human gut microbiota in early life is closely associated with both short- and long-term infant health. Delivery mode and feeding pattern are two important determinants of infant gut microbiota. In this longitudinal cohort study, we examined the interplay between the delivery mode and feeding pattern on the dynamics of infant gut microbiota from 6 weeks to 6 months post-delivery in 139 infants. We also assessed the relationship between infant respiratory infection susceptibility and gut microbial changes associated with delivery mode and feeding pattern. At 6 weeks postpartum, the composition and structure of gut microbiota of cesarean section-delivered (CSD) infants differed from those of vaginally delivered (VD) infants, with decreased Bacteroides and Escherichia-Shigella and increased Klebsiella, Veillonella, and Enterococcus. At 6 months postpartum, these delivery mode-induced microbial shifts were restored by exclusive breastfeeding, resulting in similar gut microbial profiles between VD and CSD infants who were exclusively breastfed (P = 0.57) and more variable gut microbial profiles between VD and CSD infants who were mixed fed (P < 0.001). We identified that the VD-associated genera were enriched in healthy infants, while the CSD-associated genera were enriched in infants who suffered from respiratory infections. Our findings indicate that exclusive breastfeeding may play a health-promoting role by reducing infant respiratory infection susceptibility through the restoration of gut microbiota perturbations caused by cesarean section.
Collapse
Affiliation(s)
- Yu Liu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jingmei Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Baoli Zhu
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Fei Liu
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Shengtang Qin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Na Lv
- Key Laboratory of Pathogenic Microbiology and Immunology/Institute of Microbiology, Chinese Academy of Science, Beijing, China
| | - Ye Feng
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| |
Collapse
|
218
|
Chen X, Shi Y. Determinants of microbial colonization in the premature gut. Mol Med 2023; 29:90. [PMID: 37407941 DOI: 10.1186/s10020-023-00689-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Abnormal microbial colonization in the gut at an early stage of life affects growth, development, and health, resulting in short- and long-term adverse effects. Microbial colonization patterns of preterm infants differ from those of full-term infants in that preterm babies and their mothers have more complicated prenatal and postnatal medical conditions. Maternal complications, antibiotic exposure, delivery mode, feeding type, and the use of probiotics may significantly shape the gut microbiota of preterm infants at an early stage of life; however, these influences subside with age. Although some factors and processes are difficult to intervene in or avoid, understanding the potential factors and determinants will help in developing timely strategies for a healthy gut microbiota in preterm infants. This review discusses potential determinants of gut microbial colonization in preterm infants and their underlying mechanisms.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
219
|
Rachmühl C, Lacroix C, Giorgetti A, Stoffel NU, Zimmermann MB, Brittenham GM, Geirnaert A. Validation of a batch cultivation protocol for fecal microbiota of Kenyan infants. BMC Microbiol 2023; 23:174. [PMID: 37403024 PMCID: PMC10318780 DOI: 10.1186/s12866-023-02915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The combination of cultivation studies with molecular analysis approaches allows characterization of the complex human gut microbiota in depth. In vitro cultivation studies of infants living in rural sub-Saharan Africa are scarce. In this study, a batch cultivation protocol for Kenyan infant fecal microbiota was validated. METHODS Fresh fecal samples were collected from 10 infants living in a rural area of Kenya. Samples were transported under protective conditions and subsequently prepared for inoculation within less than 30 h for batch cultivation. A diet-adapted cultivation medium was used that mimicked the daily intake of human milk and maize porridge in Kenyan infants during weaning. 16 S rRNA gene amplicon sequencing and HPLC analyses were performed to assess the composition and metabolic activity, respectively, of the fecal microbiota after 24 h of batch cultivation. RESULTS High abundance of Bifidobacterium (53.4 ± 11.1%) and high proportions of acetate (56 ± 11% of total metabolites) and lactate (24 ± 22% of total metabolites) were detected in the Kenyan infant fecal microbiota. After cultivation started at an initial pH 7.6, the fraction of top bacterial genera (≥ 1% abundant) shared between fermentation and fecal samples was high at 97 ± 5%. However, Escherichia-Shigella, Clostridium sensu stricto 1, Bacteroides and Enterococcus were enriched concomitant with decreased Bifidobacterium abundance. Decreasing the initial pH to 6.9 lead to higher abundance of Bifidobacterium after incubation and increased the compositional similarity of fermentation and fecal samples. Despite similar total metabolite production of all fecal microbiota after cultivation, inter-individual differences in metabolite profiles were apparent. CONCLUSIONS Protected transport and batch cultivation in host and diet adapted conditions allowed regrowth of the top abundant genera and reproduction of the metabolic activity of fresh Kenyan infant fecal microbiota. The validated batch cultivation protocol can be used to study the composition and functional potential of Kenyan infant fecal microbiota in vitro.
Collapse
Affiliation(s)
- Carole Rachmühl
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.
| | - Ambra Giorgetti
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Gary M Brittenham
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, USA
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.
| |
Collapse
|
220
|
Barone M, Ramayo-Caldas Y, Estellé J, Tambosco K, Chadi S, Maillard F, Gallopin M, Planchais J, Chain F, Kropp C, Rios-Covian D, Sokol H, Brigidi P, Langella P, Martín R. Gut barrier-microbiota imbalances in early life lead to higher sensitivity to inflammation in a murine model of C-section delivery. MICROBIOME 2023; 11:140. [PMID: 37394428 PMCID: PMC10316582 DOI: 10.1186/s40168-023-01584-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/25/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Most interactions between the host and its microbiota occur at the gut barrier, and primary colonizers are essential in the gut barrier maturation in the early life. The mother-offspring transmission of microorganisms is the most important factor influencing microbial colonization in mammals, and C-section delivery (CSD) is an important disruptive factor of this transfer. Recently, the deregulation of symbiotic host-microbe interactions in early life has been shown to alter the maturation of the immune system, predisposing the host to gut barrier dysfunction and inflammation. The main goal of this study is to decipher the role of the early-life gut microbiota-barrier alterations and its links with later-life risks of intestinal inflammation in a murine model of CSD. RESULTS The higher sensitivity to chemically induced inflammation in CSD mice is related to excessive exposure to a too diverse microbiota too early in life. This early microbial stimulus has short-term consequences on the host homeostasis. It switches the pup's immune response to an inflammatory context and alters the epithelium structure and the mucus-producing cells, disrupting gut homeostasis. This presence of a too diverse microbiota in the very early life involves a disproportionate short-chain fatty acids ratio and an excessive antigen exposure across the vulnerable gut barrier in the first days of life, before the gut closure. Besides, as shown by microbiota transfer experiments, the microbiota is causal in the high sensitivity of CSD mice to chemical-induced colitis and in most of the phenotypical parameters found altered in early life. Finally, supplementation with lactobacilli, the main bacterial group impacted by CSD in mice, reverts the higher sensitivity to inflammation in ex-germ-free mice colonized by CSD pups' microbiota. CONCLUSIONS Early-life gut microbiota-host crosstalk alterations related to CSD could be the linchpin behind the phenotypic effects that lead to increased susceptibility to an induced inflammation later in life in mice. Video Abstract.
Collapse
Affiliation(s)
- M. Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Y. Ramayo-Caldas
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Animal Breeding and Genetics Program, Institute for Research and Technology in Food and Agriculture (IRTA), Torre Marimon, 08140 Caldes de Montbui, Spain
| | - J. Estellé
- INRAE, AgroParisTech, GABI, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - K. Tambosco
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - S. Chadi
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Maillard
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - M. Gallopin
- CNRS, CEA, l’Institut de Biologie Intégrative de La Cellule (I2BC), Paris-Saclay University, 91405 Orsay, France
| | - J. Planchais
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - F. Chain
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - C. Kropp
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - D. Rios-Covian
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - H. Sokol
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Centre de Recherche Saint-Antoine, CRSA, AP-HP, INSERM, Saint Antoine Hospital, Sorbonne Université, 75012 Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - P. Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - P. Langella
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - R. Martín
- INRAE, AgroParisTech, Micalis Institut,, Paris-Saclay University, 78350 Jouy-en-Josas, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| |
Collapse
|
221
|
Kindgren E, Ahrens AP, Triplett EW, Ludvigsson J. Infant gut microbiota and environment associate with juvenile idiopathic arthritis many years prior to disease onset, especially in genetically vulnerable children. EBioMedicine 2023; 93:104654. [PMID: 37329576 PMCID: PMC10279551 DOI: 10.1016/j.ebiom.2023.104654] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/13/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND The etiology of juvenile idiopathic arthritis (JIA) is poorly understood. This study investigated genetic and environmental factors and infant gut microbiota in a prospective birth cohort to assess disease risk. METHODS Data was collected from the All Babies in Southeast Sweden (ABIS) population-based cohort (n = 17,055), 111 of whom later acquired JIA (ABISJIA). Stool samples were collected at one year of age for 10.4%. To determine disease association, 16S rRNA gene sequences were analyzed, with and without confound adjustment. Genetic and environmental risks were assessed. FINDINGS ABISJIA had higher abundance of Acidaminococcales, Prevotella 9, and Veillonella parvula and lower abundance of Coprococcus, Subdoligranulum, Phascolarctobacterium, Dialister spp., Bifidobacterium breve, Fusicatenibacter saccharivorans, Roseburia intestinalis, and Akkermansia muciniphila (q's < 0.05). Parabacteroides distasonis greatly increased the odds of later contracting JIA (OR = 6.7; 1.81-24.84, p = 0.0045). Shorter breastfeeding duration and increased antibiotic exposure compounded risk in a dose-dependent manner, especially in those with genetic predisposition. INTERPRETATION Microbial dysregulation in infancy may trigger or accelerate JIA development. Environmental risk factors have a stronger impact on genetically predisposed children. This study is the first to implicate microbial dysregulation in JIA at such an early age, with many bacterial taxa associated with risk factors. These findings provide opportunities for intervention or early screening and offer new insights into JIA pathogenesis. FUNDING Barndiabetesfonden; Swedish Council for Working Life and Social Research; Swedish Research Council; Östgöta Brandstodsbolag; Medical Research Council of Southeast Sweden; JDRF-Wallenberg Foundation; Linköping.
Collapse
Affiliation(s)
- Erik Kindgren
- Department of Pediatrics, Region Västra Götaland, Skaraborg Hospital, Skövde, Sweden; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Angelica P Ahrens
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611-0700, USA
| | - Eric W Triplett
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611-0700, USA.
| | - Johnny Ludvigsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Crown Princess Victoria's Children's Hospital, Region Östergötland, Linköping, SE 58185, Sweden
| |
Collapse
|
222
|
Mueller NT, Differding MK, Sun H, Wang J, Levy S, Deopujari V, Appel LJ, Blaser MJ, Kundu T, Shah AA, Dominguez Bello MG, Hourigan SK. Maternal Bacterial Engraftment in Multiple Body Sites of Cesarean Section Born Neonates after Vaginal Seeding-a Randomized Controlled Trial. mBio 2023; 14:e0049123. [PMID: 37074174 PMCID: PMC10294643 DOI: 10.1128/mbio.00491-23] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023] Open
Abstract
Children delivered by elective, prelabor Cesarean section (C-section) are not exposed to the birth canal microbiota and, in relation to vaginally delivered children, show altered microbiota development. Perturbed microbial colonization during critical early-life windows of development alters metabolic and immune programming and is associated with an increased risk of immune and metabolic diseases. In nonrandomized studies, vaginal seeding of C-section-born neonates partially restores their microbiota colonization to that of their vaginally delivered counterparts, but without randomization, confounding factors cannot be excluded. In a double-blind, randomized, placebo-controlled trial, we determined the effect of vaginal seeding versus placebo seeding (control arm) on the skin and stool microbiota of elective, prelabor C-section-born neonates (n = 20) at 1 day and 1 month after birth. We also examined whether there were between-arm differences in engraftment of maternal microbes in the neonatal microbiota. In relation to the control arm, vaginal seeding increased mother-to-neonate microbiota transmission and caused compositional changes and a reduction in alpha diversity (Shannon Index) of the skin and stool microbiota. The neonatal skin and stool microbiota alpha diversity when maternal vaginal microbiota is provided is intriguing and highlights the need of larger randomized studies to determine the ecological mechanisms and effects of vaginal seeding on clinical outcomes. IMPORTANCE Children delivered by elective C-section are not exposed to the birth canal and show altered microbiota development. Impairing microbial colonization during early life alters metabolic and immune programming and is associated with an increased risk of immune and metabolic diseases. In a double-blind, randomized, placebo-controlled trial, we determined the effect of vaginal seeding on the skin and stool microbiota of elective C-section born neonates and found that vaginal seeding increased mother-to-neonate microbiota transmission and caused compositional changes and a reduction in the skin and stool microbiota diversity. The reduction of neonatal skin and stool microbiota diversity when maternal vaginal microbiota is provided is intriguing and highlights the need of larger randomized studies to determine the ecological mechanisms and effects of vaginal seeding on clinical outcomes.
Collapse
Affiliation(s)
- Noel T. Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Moira K. Differding
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Haipeng Sun
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Jincheng Wang
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Shira Levy
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Varsha Deopujari
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Lawrence J. Appel
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, USA
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, New Jersey, USA
| | - Tanima Kundu
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, New Jersey, USA
| | - Ankit A. Shah
- Inova Women’s Hospital, Inova Health System, Falls Church, Virginia, USA
| | - Maria Gloria Dominguez Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Anthropology, Rutgers University, New Brunswick, New Jersey, USA
- Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, New Jersey, USA
- Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| | - Suchitra K. Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Inova Children’s Hospital, Inova Health System, Falls Church, Virginia, USA
| |
Collapse
|
223
|
Bakshi S, Paswan VK, Yadav SP, Bhinchhar BK, Kharkwal S, Rose H, Kanetkar P, Kumar V, Al-Zamani ZAS, Bunkar DS. A comprehensive review on infant formula: nutritional and functional constituents, recent trends in processing and its impact on infants' gut microbiota. Front Nutr 2023; 10:1194679. [PMID: 37415910 PMCID: PMC10320619 DOI: 10.3389/fnut.2023.1194679] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 07/08/2023] Open
Abstract
Human milk is considered the most valuable form of nutrition for infants for their growth, development and function. So far, there are still some cases where feeding human milk is not feasible. As a result, the market for infant formula is widely increasing, and formula feeding become an alternative or substitute for breastfeeding. The nutritional value of the formula can be improved by adding functional bioactive compounds like probiotics, prebiotics, human milk oligosaccharides, vitamins, minerals, taurine, inositol, osteopontin, lactoferrin, gangliosides, carnitine etc. For processing of infant formula, diverse thermal and non-thermal technologies have been employed. Infant formula can be either in powdered form, which requires reconstitution with water or in ready-to-feed liquid form, among which powder form is readily available, shelf-stable and vastly marketed. Infants' gut microbiota is a complex ecosystem and the nutrient composition of infant formula is recognized to have a lasting effect on it. Likewise, the gut microbiota establishment closely parallels with host immune development and growth. Therefore, it must be contemplated as an important factor for consideration while developing formulas. In this review, we have focused on the formulation and manufacturing of safe and nutritious infant formula equivalent to human milk or aligning with the infant's needs and its ultimate impact on infants' gut microbiota.
Collapse
Affiliation(s)
- Shiva Bakshi
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Vinod Kumar Paswan
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Satya Prakash Yadav
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Basant Kumar Bhinchhar
- Department of Livestock Production Management, Sri Karan Narendra Agriculture University, Jobner, India
| | - Sheela Kharkwal
- Department of Agriculture Economics, Sri Karan Narendra Agriculture University, Jobner, India
| | - Hency Rose
- Division of Dairy Technology, ICAR—National Dairy Research Institute, Karnal, India
| | - Prajasattak Kanetkar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Zakarya Ali Saleh Al-Zamani
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
- Department of Food Technology and Science, Faculty of Agriculture and Veterinary Medicine, Ibb University, Ibb, Yemen
| | - Durga Shankar Bunkar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
224
|
Zhang J, Zhang C, Zhang Q, Yu L, Chen W, Xue Y, Zhai Q. Meta-analysis of the effects of proton pump inhibitors on the human gut microbiota. BMC Microbiol 2023; 23:171. [PMID: 37337143 DOI: 10.1186/s12866-023-02895-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023] Open
Abstract
Mounting evidence has linked changes in human gut microbiota to proton pump inhibitor (PPI) use. Accordingly, multiple studies have analyzed the gut microbiomes of PPI users, but PPI-microbe interactions are still understudied. Here, we performed a meta-analysis of four studies with available 16S rRNA gene amplicon sequencing data to uncover the potential changes in human gut microbes among PPI users. Despite some differences, we found common features of the PPI-specific microbiota, including a decrease in the Shannon diversity index and the depletion of bacteria from the Ruminococcaceae and Lachnospiraceae families, which are crucial short-chain fatty acid-producers. Through training based on multiple studies, using a random forest classification model, we further verified the representativeness of the six screened gut microbial genera and 20 functional genes as PPI-related biomarkers, with AUC values of 0.748 and 0.879, respectively. Functional analysis of the PPI-associated 16S rRNA microbiome revealed enriched carbohydrate- and energy-associated genes, mostly encoding fructose-1,6-bisphosphatase and pyruvate dehydrogenase, among others. In this study, we have demonstrated alterations in bacterial abundance and functional metabolic potential related to PPI use, as a basis for future studies on PPI-induced adverse effects.
Collapse
Affiliation(s)
- Jiayi Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Qingsong Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Jiangsu Province, Wuxi, China.
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, People's Republic of China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
225
|
Rusch JA, Layden BT, Dugas LR. Signalling cognition: the gut microbiota and hypothalamic-pituitary-adrenal axis. Front Endocrinol (Lausanne) 2023; 14:1130689. [PMID: 37404311 PMCID: PMC10316519 DOI: 10.3389/fendo.2023.1130689] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/25/2023] [Indexed: 07/06/2023] Open
Abstract
Cognitive function in humans depends on the complex and interplay between multiple body systems, including the hypothalamic-pituitary-adrenal (HPA) axis. The gut microbiota, which vastly outnumbers human cells and has a genetic potential that exceeds that of the human genome, plays a crucial role in this interplay. The microbiota-gut-brain (MGB) axis is a bidirectional signalling pathway that operates through neural, endocrine, immune, and metabolic pathways. One of the major neuroendocrine systems responding to stress is the HPA axis which produces glucocorticoids such as cortisol in humans and corticosterone in rodents. Appropriate concentrations of cortisol are essential for normal neurodevelopment and function, as well as cognitive processes such as learning and memory, and studies have shown that microbes modulate the HPA axis throughout life. Stress can significantly impact the MGB axis via the HPA axis and other pathways. Animal research has advanced our understanding of these mechanisms and pathways, leading to a paradigm shift in conceptual thinking about the influence of the microbiota on human health and disease. Preclinical and human trials are currently underway to determine how these animal models translate to humans. In this review article, we summarize the current knowledge of the relationship between the gut microbiota, HPA axis, and cognition, and provide an overview of the main findings and conclusions in this broad field.
Collapse
Affiliation(s)
- Jody A. Rusch
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- C17 Chemical Pathology Laboratory, Groote Schuur Hospital, National Health Laboratory Service, Cape Town, South Africa
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Lara R. Dugas
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
226
|
Li J, Li D, Chen Y, Chen W, Xu J, Gao L. Gut Microbiota and Aging: Traditional Chinese Medicine and Modern Medicine. Clin Interv Aging 2023; 18:963-986. [PMID: 37351381 PMCID: PMC10284159 DOI: 10.2147/cia.s414714] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 06/24/2023] Open
Abstract
The changing composition of gut microbiota, much like aging, accompanies people throughout their lives, and the inextricable relationship between both has recently attracted extensive attention as well. Modern medical research has revealed that a series of changes in gut microbiota are involved in the aging process of organisms, which may be because gut microbiota modulates aging-related changes related to innate immunity and cognitive function. At present, there is no definite and effective method to delay aging. However, Nobel laureate Tu Youyou's research on artemisinin has inspired researchers to study the importance of Traditional Chinese Medicine (TCM). TCM, as an ancient alternative medicine, has unique advantages in preventive health care and in treating diseases as it already has formed an independent understanding of the aging system. TCM practitioners believe that the mechanism of aging is mainly deficiency, and pathological states such as blood stasis, qi stagnation and phlegm coagulation can exacerbate the process of aging, which involves a series of organs, including the brain, kidney, heart, liver and spleen. Our current understanding of aging has led us to realise that TCM can indeed make some beneficial changes, such as the improvement of cognitive impairment. However, due to the multi-component and multi-target nature of TCM, the exploration of its mechanism of action has become extremely complex. While analysing the relationship between gut microbiota and aging, this review explores the similarities and differences in treatment methods and mechanisms between TCM and Modern Medicine, in order to explore a new approach that combines TCM and Modern Medicine to regulate gut microbiota, improve immunity and delay aging.
Collapse
Affiliation(s)
- Jinfan Li
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, People’s Republic of China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Dong Li
- Department of Diabetes, Licheng District Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250100, People’s Republic of China
| | - Yajie Chen
- Department of Rehabilitation and Health Care, Jinan Vocational College of Nursing, Jinan, Shandong, 250100, People’s Republic of China
| | - Wenbin Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| |
Collapse
|
227
|
Borbet TC, Pawline MB, Li J, Ho ML, Yin YS, Zhang X, Novikova E, Jackson K, Mullins BJ, Ruiz VE, Hines MJ, Zhang XS, Müller A, Koralov SB, Blaser MJ. Disruption of the early-life microbiota alters Peyer's patch development and germinal center formation in gastrointestinal-associated lymphoid tissue. iScience 2023; 26:106810. [PMID: 37235047 PMCID: PMC10206152 DOI: 10.1016/j.isci.2023.106810] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/17/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
During postnatal development, both the maturing microbiome and the host immune system are susceptible to environmental perturbations such as antibiotic use. The impact of timing in which antibiotic exposure occurs was investigated by treating mice from days 5-9 with amoxicillin or azithromycin, two of the most commonly prescribed medications in children. Both early-life antibiotic regimens disrupted Peyer's patch development and immune cell abundance, with a sustained decrease in germinal center formation and diminished intestinal immunoglobulin A (IgA) production. These effects were less pronounced in adult mice. Through comparative analysis of microbial taxa, Bifidobacterium longum abundance was found to be associated with germinal center frequency. When re-introduced to antibiotic-exposed mice, B. longum partially rescued the immunological deficits. These findings suggest that early-life antibiotic use affects the development of intestinal IgA-producing B cell functions and that probiotic strains could be used to restore normal development after antibiotic exposure.
Collapse
Affiliation(s)
- Timothy C. Borbet
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Miranda B. Pawline
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Jackie Li
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Melody L. Ho
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Yue Sandra Yin
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| | - Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Ekaterina Novikova
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Katelyn Jackson
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Briana J. Mullins
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Victoria E. Ruiz
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Marcus J. Hines
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Xue-Song Zhang
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Sergei B. Koralov
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Martin J. Blaser
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| |
Collapse
|
228
|
McMath AL, Aguilar-Lopez M, Cannavale CN, Khan NA, Donovan SM. A systematic review on the impact of gastrointestinal microbiota composition and function on cognition in healthy infants and children. Front Neurosci 2023; 17:1171970. [PMID: 37389363 PMCID: PMC10306408 DOI: 10.3389/fnins.2023.1171970] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Evidence from animal models or children with neurodevelopmental disorders has implicated the gut microbiome (GM) in neurocognitive development. However, even subclinical impairement of cognition can have negative consequences, as cognition serves as the foundation for skills necessary to succeed in school, vocation and socially. The present study aims to identify gut microbiome characteristics or changes in gut microbiome characteristics that consistently associate with cognitive outcomes in healthy, neurotypical infants and children. Of the 1,520 articles identified in the search, 23 were included in qualitative synthesis after applying exclusion criteria. Most studies were cross-sectional and focused on behavior or motor and language skills. Bifidobacterium, Bacteroides, Clostridia, Prevotella, and Roseburia were related to these aspects of cognition across several studies. While these results support the role of GM in cognitive development, higher quality studies focused on more complex cognition are needed to understand the extent to which the GM contributes to cognitive development.
Collapse
Affiliation(s)
- Arden L. McMath
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Miriam Aguilar-Lopez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Corinne N. Cannavale
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Naiman A. Khan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois Urbana-Champaign, Champaign, IL, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
229
|
Shin H, Martinez KA, Henderson N, Jay M, Schweizer W, Bogaert D, Park G, Bokulich NA, Blaser MJ, Dominguez-Bello MG. Partial convergence of the human vaginal and rectal maternal microbiota in late gestation and early post-partum. NPJ Biofilms Microbiomes 2023; 9:37. [PMID: 37311781 PMCID: PMC10264455 DOI: 10.1038/s41522-023-00404-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 06/02/2023] [Indexed: 06/15/2023] Open
Abstract
The human vaginal and fecal microbiota change during pregnancy. Because of the proximity of these perineal sites and the evolutionarily conserved maternal-to-neonatal transmission of the microbiota, we hypothesized that the microbiota of these two sites (rectal and vaginal) converge during the last gestational trimester as part of the preparation for parturition. To test this hypothesis, we analyzed 16S rRNA sequences from vaginal introitus and rectal samples in 41 women at gestational ages 6 and 8 months, and at 2 months post-partum. The results show that the human vaginal and rectal bacterial microbiota converged during the last gestational trimester and into the 2nd month after birth, with a significant decrease in Lactobacillus species in both sites, as alpha diversity progressively increased in the vagina and decreased in the rectum. The microbiota convergence of the maternal vaginal-anal sites perinatally might hold significance for the inter-generational transmission of the maternal microbiota.
Collapse
Affiliation(s)
- Hakdong Shin
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, South Korea
| | - Keith A Martinez
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA
| | - Nora Henderson
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
| | - Melanie Jay
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
- Department of Population Health, New York University Langone Medical Center, New York, NY, USA
| | - William Schweizer
- Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Debby Bogaert
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, Scotland
| | - Gwoncheol Park
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, South Korea
| | - Nicholas A Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA.
- Department of Anthropology, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
230
|
Kozhakhmetov S, Meiirmanova Z, Mukhanbetzhanov N, Jarmukhanov Z, Vinogradova E, Mureyev S, Kozhakhmetova S, Morenko M, Shnaider K, Duisbayeva A, Kushugulova A. Compositional and functional variability of the gut microbiome in children with infantile colic. Sci Rep 2023; 13:9530. [PMID: 37308527 DOI: 10.1038/s41598-023-36641-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023] Open
Abstract
The inconsolable crying of a child for no apparent reason at an early age is a source of excitement and anxiety for parents. Previous studies have reported that crying may be caused by discomfort associated with the occupation of the intestines of the newborn by microbiota and its vital activity. We conducted a prospective observational study in which 62 newborns and their mothers were recruited. The study comprised two groups, each consisting of 15 infants with colic and 21 controls. Colic and control groups were vaginally born and exclusively breastfed. Fecal samples from children were collected over time from day 1 to 12 months. Full metagenomic sequencing of fecal samples from children and their mothers was carried out. It was determined that the trajectory of the development of the intestinal microbiome of children with colic was different from the group without colic. In the colic group, a depleted relative abundance of Bifidobacterium and enrichment of Bacteroides Clostridiales was found, while the microbial biodiversity in this group was enriched. Metabolic pathway profiling showed that the non-colic group was enriched by amino acid biosynthetic pathways, while the feces microbiome of the colic group was enriched by glycolysis metabolic pathways that correlated with the Bacteroides taxon. This study shows that infantile colic has a definite relationship with the microbiome structure of infants.
Collapse
Affiliation(s)
- Samat Kozhakhmetov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan.
| | - Zarina Meiirmanova
- Department of Children's Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC "Astana Medical University", Astana, Z01G6C5, Kazakhstan
| | - Nurislam Mukhanbetzhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan
| | - Zharkyn Jarmukhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan
| | - Elizaveta Vinogradova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan
| | - Shamil Mureyev
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan
| | | | - Marina Morenko
- Department of Children's Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC "Astana Medical University", Astana, Z01G6C5, Kazakhstan
| | - Kseniya Shnaider
- Department of Children's Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC "Astana Medical University", Astana, Z01G6C5, Kazakhstan
| | - Arailym Duisbayeva
- Department of Children's Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC "Astana Medical University", Astana, Z01G6C5, Kazakhstan
| | - Almagul Kushugulova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr ave., Block S1, Nur-Sultan, Z05H0P9, Republic of Kazakhstan.
| |
Collapse
|
231
|
Yi X, Zhou K, Jiang P, Deng N, Peng X, Tan Z. Brain-bacteria-gut axis and oxidative stress mediated by intestinal mucosal microbiota might be an important mechanism for constipation in mice. 3 Biotech 2023; 13:192. [PMID: 37205176 PMCID: PMC10185723 DOI: 10.1007/s13205-023-03580-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Intestinal microbiota disorder was associated with constipation. This study investigated the microbiota-gut-brain axis and oxidative stress mediated by intestinal mucosal microbiota in mice with spleen deficiency constipation. The Kunming mice were randomly divided into the control (MC) group and the constipation (MM) group. The spleen deficiency constipation model was established by gavage with Folium sennae decoction and controlled diet and water intake. The body weight, spleen and thymus index, 5-Hydroxytryptamine (5-HT) and Superoxide Dismutase (SOD) content were significantly lower in the MM group than the MC group, the content of vasoactive intestinal peptide (VIP) and malondialdehyde (MDA) content were significantly higher than the MC group. The Alpha diversity of intestinal mucosal bacteria was not changed but beta diversity was changed in mice with spleen deficiency constipation. Compared to the MC group, the relative abundance of Proteobacteria was an upward trend and the Firmicutes/Bacteroidota (F/B) value was a downward trend in the MM group. There was a significant difference in the characteristic microbiota between the two groups. In the MM group, Brevinema, Akkermansia, Parasutterella, Faecalibaculum, Aeromonas, Sphingobium, Actinobacillus, and other pathogenic bacteria were enriched. Meanwhile, there was a certain relationship between the microbiota and gastrointestinal neuropeptide and oxidative stress indicators. The community structure of intestinal mucosal bacteria in mice with spleen deficiency constipation was changed, which was characterized by the reduction of F/B value and enrichment of Proteobacteria. Microbiota-gut-brain axis may be important for spleen deficiency constipation.
Collapse
Affiliation(s)
- Xin Yi
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Kang Zhou
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Ping Jiang
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007 China
| | - Na Deng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| | - Xinxin Peng
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007 China
| | - Zhoujin Tan
- The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208 China
| |
Collapse
|
232
|
Abstract
Antibiotic therapy remains a cornerstone of treatment of both medical and surgical presentations of necrotizing enterocolitis (NEC). However, guidelines regarding the administration of antibiotics for the treatment of NEC are lacking and practices vary amongst clinicians. Although the pathogenesis of NEC is unknown, there is consensus that the infant gastrointestinal microbiome contributes to the disease. The presumed connection between dysbiosis and NEC has prompted some to study whether early prophylactic enteral antibiotics can prevent NEC. Yet others have taken an opposing approach, studying whether perinatal antibiotic exposure increases the risk of NEC by inducing a state of dysbiosis. This narrative review summarizes what is known about antibiotics and their association with the infant microbiome and NEC, current antibiotic prescribing practices for infants with medical and surgical NEC, as well as potential strategies to further optimize the use of antibiotics in this population of infants.
Collapse
Affiliation(s)
- Elizabeth Pace
- University of Pittsburgh Department of Surgery, United States
| | - Toby D Yanowitz
- University of Pittsburgh Department of Pediatrics, Division of Neonatology, United States
| | - Paul Waltz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States
| | - Michael J Morowitz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States.
| |
Collapse
|
233
|
Ormaasen I, Rudi K, Diep DB, Snipen L. Metagenome-mining indicates an association between bacteriocin presence and strain diversity in the infant gut. BMC Genomics 2023; 24:295. [PMID: 37259063 PMCID: PMC10230729 DOI: 10.1186/s12864-023-09388-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Our knowledge about the ecological role of bacterial antimicrobial peptides (bacteriocins) in the human gut is limited, particularly in relation to their role in the diversification of the gut microbiota during early life. The aim of this paper was therefore to address associations between bacteriocins and bacterial diversity in the human gut microbiota. To investigate this, we did an extensive screening of 2564 healthy human gut metagenomes for the presence of predicted bacteriocin-encoding genes, comparing bacteriocin gene presence to strain diversity and age. RESULTS We found that the abundance of bacteriocin genes was significantly higher in infant-like metagenomes (< 2 years) compared to adult-like metagenomes (2-107 years). By comparing infant-like metagenomes with and without a given bacteriocin, we found that bacteriocin presence was associated with increased strain diversities. CONCLUSIONS Our findings indicate that bacteriocins may play a role in the strain diversification during the infant gut microbiota establishment.
Collapse
Affiliation(s)
- Ida Ormaasen
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway.
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Dzung B Diep
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Lars Snipen
- Faculty of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
234
|
Manara S, Selma-Royo M, Huang KD, Asnicar F, Armanini F, Blanco-Miguez A, Cumbo F, Golzato D, Manghi P, Pinto F, Valles-Colomer M, Amoroso L, Corrias MV, Ponzoni M, Raffaetà R, Cabrera-Rubio R, Olcina M, Pasolli E, Collado MC, Segata N. Maternal and food microbial sources shape the infant microbiome of a rural Ethiopian population. Curr Biol 2023; 33:1939-1950.e4. [PMID: 37116481 PMCID: PMC10234599 DOI: 10.1016/j.cub.2023.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 02/28/2023] [Accepted: 04/05/2023] [Indexed: 04/30/2023]
Abstract
The human microbiome seeding starts at birth, when pioneer microbes are acquired mainly from the mother. Mode of delivery, antibiotic prophylaxis, and feeding method have been studied as modulators of mother-to-infant microbiome transmission, but other key influencing factors like modern westernized lifestyles with high hygienization, high-calorie diets, and urban settings, compared with non-westernized lifestyles have not been investigated yet. In this study, we explored the mother-infant sharing of characterized and uncharacterized microbiome members via strain-resolved metagenomics in a cohort of Ethiopian mothers and infants, and we compared them with four other cohorts with different lifestyles. The westernized and non-westernized newborns' microbiomes composition overlapped during the first months of life more than later in life, likely reflecting similar initial breast-milk-based diets. Ethiopian and other non-westernized infants shared a smaller fraction of the microbiome with their mothers than did most westernized populations, despite showing a higher microbiome diversity, and uncharacterized species represented a substantial fraction of those shared in the Ethiopian cohort. Moreover, we identified uncharacterized species belonging to the Selenomonadaceae and Prevotellaceae families specifically present and shared only in the Ethiopian cohort, and we showed that a locally produced fermented food, injera, can contribute to the higher diversity observed in the Ethiopian infants' gut with bacteria that are not part of the human microbiome but are acquired through fermented food consumption. Taken together, these findings highlight the fact that lifestyle can impact the gut microbiome composition not only through differences in diet, drug consumption, and environmental factors but also through its effect on mother-infant strain-sharing patterns.
Collapse
Affiliation(s)
- Serena Manara
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Marta Selma-Royo
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy; Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), C/ Catedrático Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| | - Kun D Huang
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Francesco Asnicar
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Federica Armanini
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Aitor Blanco-Miguez
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Fabio Cumbo
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Davide Golzato
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Paolo Manghi
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Federica Pinto
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Mireia Valles-Colomer
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy
| | - Loredana Amoroso
- Oncology Unit, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Roberta Raffaetà
- Ca' Foscari University Venice, Department of Philosophy and Cultural Heritage and NICHE, Malcanton Marcorà, Dorsoduro 3484/D, 30123 Venice, Italy
| | - Raul Cabrera-Rubio
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), C/ Catedrático Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain
| | - Mari Olcina
- Department of Preventive Medicine and Public Health, Faculty of Pharmacy, Universitat de València, Av. Vicent Andrés Estellés s/n, Burjassot, Valencia 46100, Spain
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Via Università 100, 80055 Portici, Naples, Italy.
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), C/ Catedrático Agustín Escardino Benlloch, 7, 46980 Paterna, Valencia, Spain.
| | - Nicola Segata
- Department of Cellular Computational and Integrative Biology, Via Sommarive 9, Povo, Trento 38123, Italy.
| |
Collapse
|
235
|
Fung DLX, Li X, Leung CK, Hu P. A self-knowledge distillation-driven CNN-LSTM model for predicting disease outcomes using longitudinal microbiome data. BIOINFORMATICS ADVANCES 2023; 3:vbad059. [PMID: 37228387 PMCID: PMC10203376 DOI: 10.1093/bioadv/vbad059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023]
Abstract
Motivation Human microbiome is complex and highly dynamic in nature. Dynamic patterns of the microbiome can capture more information than single point inference as it contains the temporal changes information. However, dynamic information of the human microbiome can be hard to be captured due to the complexity of obtaining the longitudinal data with a large volume of missing data that in conjunction with heterogeneity may provide a challenge for the data analysis. Results We propose using an efficient hybrid deep learning architecture convolutional neural network-long short-term memory, which combines with self-knowledge distillation to create highly accurate models to analyze the longitudinal microbiome profiles to predict disease outcomes. Using our proposed models, we analyzed the datasets from Predicting Response to Standardized Pediatric Colitis Therapy (PROTECT) study and DIABIMMUNE study. We showed the significant improvement in the area under the receiver operating characteristic curve scores, achieving 0.889 and 0.798 on PROTECT study and DIABIMMUNE study, respectively, compared with state-of-the-art temporal deep learning models. Our findings provide an effective artificial intelligence-based tool to predict disease outcomes using longitudinal microbiome profiles from collected patients. Availability and implementation The data and source code can be accessed at https://github.com/darylfung96/UC-disease-TL.
Collapse
Affiliation(s)
- Daryl L X Fung
- Department of Computer Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Xu Li
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Carson K Leung
- Department of Computer Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | |
Collapse
|
236
|
Martin AJ, Serebrinsky-Duek K, Riquelme E, Saa PA, Garrido D. Microbial interactions and the homeostasis of the gut microbiome: the role of Bifidobacterium. MICROBIOME RESEARCH REPORTS 2023; 2:17. [PMID: 38046822 PMCID: PMC10688804 DOI: 10.20517/mrr.2023.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 12/05/2023]
Abstract
The human gut is home to trillions of microorganisms that influence several aspects of our health. This dense microbial community targets almost all dietary polysaccharides and releases multiple metabolites, some of which have physiological effects on the host. A healthy equilibrium between members of the gut microbiota, its microbial diversity, and their metabolites is required for intestinal health, promoting regulatory or anti-inflammatory immune responses. In contrast, the loss of this equilibrium due to antibiotics, low fiber intake, or other conditions results in alterations in gut microbiota composition, a term known as gut dysbiosis. This dysbiosis can be characterized by a reduction in health-associated microorganisms, such as butyrate-producing bacteria, enrichment of a small number of opportunistic pathogens, or a reduction in microbial diversity. Bifidobacterium species are key species in the gut microbiome, serving as primary degraders and contributing to a balanced gut environment in various ways. Colonization resistance is a fundamental property of gut microbiota for the prevention and control of infections. This community competes strongly with foreign microorganisms, such as gastrointestinal pathogens, antibiotic-resistant bacteria, or even probiotics. Resistance to colonization is based on microbial interactions such as metabolic cross-feeding, competition for nutrients, or antimicrobial-based inhibition. These interactions are mediated by metabolites and metabolic pathways, representing the inner workings of the gut microbiota, and play a protective role through colonization resistance. This review presents a rationale for how microbial interactions provide resistance to colonization and gut dysbiosis, highlighting the protective role of Bifidobacterium species.
Collapse
Affiliation(s)
- Alberto J.M. Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago 8580702, Chile
| | - Kineret Serebrinsky-Duek
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| | - Erick Riquelme
- Department of Respiratory Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Pedro A. Saa
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
- Institute for Mathematical and Computational Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, Pontificia Universidad Católica de Chile, Santiago 833115, Chile
| |
Collapse
|
237
|
Thomas SP, Denizer E, Zuffa S, Best BM, Bode L, Chambers CD, Dorrestein PC, Liu GY, Momper JD, Nizet V, Tsunoda SM, Tremoulet AH. Transfer of antibiotics and their metabolites in human milk: Implications for infant health and microbiota. Pharmacotherapy 2023; 43:442-451. [PMID: 36181712 PMCID: PMC10763576 DOI: 10.1002/phar.2732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 05/17/2023]
Abstract
Antibiotics are an essential tool for perinatal care. While antibiotics can play a life-saving role for both parents and infants, they also cause collateral damage to the beneficial bacteria that make up the host gut microbiota. This is especially true for infants, whose developing gut microbiota is uniquely sensitive to antibiotic perturbation. Emerging evidence suggests that disruption of these bacterial populations during this crucial developmental window can have long-term effects on infant health and development. Although most current studies have focused on microbial disruptions caused by direct antibiotic administration to infants or prenatal exposure to antibiotics administered to the mother, little is known about whether antibiotics in human milk may pose similar risks to the infant. This review surveys current data on antibiotic transfer during lactation and highlights new methodologies to assess drug transfer in human milk. Finally, we provide recommendations for future work to ensure antibiotic use in lactating parents is safe and effective for both parents and infants.
Collapse
Affiliation(s)
- Sydney P. Thomas
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, UC San Diego, La Jolla, California, USA
| | - Erce Denizer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, UC San Diego, La Jolla, California, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, UC San Diego, La Jolla, California, USA
| | - Brookie M. Best
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
| | - Lars Bode
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
- Mother-Milk-Infant Center of Research Excellence (MOMI CORE), UC San Diego, La Jolla, California, USA
| | - Christina D. Chambers
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
- Hebert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, California, USA
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, UC San Diego, La Jolla, California, USA
| | - George Y. Liu
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
| | - Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Victor Nizet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
| | - Shirley M. Tsunoda
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Adriana H. Tremoulet
- Pediatrics Department-Rady Children's Hospital San Diego, UC San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
238
|
Kedia S, Ahuja V. Human gut microbiome: A primer for the clinician. JGH Open 2023; 7:337-350. [PMID: 37265934 PMCID: PMC10230107 DOI: 10.1002/jgh3.12902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/14/2022] [Accepted: 04/01/2023] [Indexed: 06/03/2023]
Abstract
The human host gets tremendously influenced by a genetically and phenotypically distinct and heterogeneous constellation of microbial species-the human microbiome-the gut being one of the most densely populated and characterized site for these organisms. Microbiome science has advanced rapidly, technically with respect to the analytical methods and biologically with respect to its mechanistic influence in health and disease states. A clinician conducting a microbiome study should be aware of the nuances related to microbiome research, especially with respect to the technical and biological factors that can influence the interpretation of research outcomes. Hence, this review is an attempt to detail these aspects of the human gut microbiome, with emphasis on its determinants in a healthy state.
Collapse
Affiliation(s)
- Saurabh Kedia
- Department of GastroenterologyAll India Institute of Medical SciencesNew DelhiIndia
| | - Vineet Ahuja
- Department of GastroenterologyAll India Institute of Medical SciencesNew DelhiIndia
| |
Collapse
|
239
|
Zyoud SH, Shakhshir M, Abushanab AS, Koni A, Taha AA, Abushamma F, Sabateen A, Al-Jabi SW. Global trends in research related to the links between microbiota and antibiotics: a visualization study. Sci Rep 2023; 13:6890. [PMID: 37106254 PMCID: PMC10140037 DOI: 10.1038/s41598-023-34187-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023] Open
Abstract
The scientific community widely acknowledges that the gut microbiota plays a critical role in maintaining host health and can be altered by a range of factors, such as antibiotic use, diet, stress, and infections. Therefore, this study utilized bibliometric analysis to thoroughly investigate research trends in the microbiota and antibiotics. Scopus was used to extract papers linked to microbiota and antibiotics published between 2002 and 2021, and both Microsoft Excel and VOSviewer were used to conduct the analysis of the data. A total of 2,816 publications discussed the connection between the microbiota and antibiotics. Growth occurred in two stages: the first (2002-2015) was characterized by fairly slow publication production, while the second (2016-2021) saw a rapid increase in publishing progress. The United States has the most publications, 654, representing 23.22% of the total. China came second with 372 publications (13.21%), followed by the United Kingdom with 161 publications (5.72%) and India with 157 publications (5.58%). In addition, publications on 'altered intestinal microbiota composition with antibiotic treatment' were introduced after 2017, while 'gut microbiota and antimicrobial resistance' and 'probiotics as an alternative antimicrobial therapy' were introduced before 2017. Based on these results, this study provides an in-depth look at key moments in the history of microbiota and antibiotic research, as well as possible directions for future research in different areas of microbiota and antibiotic research. Therefore, it is suggested that more attention should be given to the latest promising hotspots, such as how antibiotic treatment changes the composition of the gut microbiota.
Collapse
Affiliation(s)
- Sa'ed H Zyoud
- Poison Control and Drug Information Center (PCDIC), College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Clinical Research Centre, An-Najah National University Hospital, Nablus, 44839, Palestine.
| | - Muna Shakhshir
- Department of Nutrition, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Amani S Abushanab
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine
| | - Amer Koni
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine
- Division of Clinical Pharmacy, Hematology and Oncology Pharmacy Department, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Adham Abu Taha
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine.
- Department of Pathology, An-Najah National University Hospital, Nablus, 44839, Palestine.
| | - Faris Abushamma
- Department of Medicine, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine
- Department of Urology, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Ali Sabateen
- Infection Control Department, An-Najah National University Hospital, Nablus, 44839, Palestine
| | - Samah W Al-Jabi
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus, 44839, Palestine
| |
Collapse
|
240
|
Mady EA, Doghish AS, El-Dakroury WA, Elkhawaga SY, Ismail A, El-Mahdy HA, Elsakka EGE, El-Husseiny HM. Impact of the mother's gut microbiota on infant microbiome and brain development. Neurosci Biobehav Rev 2023; 150:105195. [PMID: 37100161 DOI: 10.1016/j.neubiorev.2023.105195] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023]
Abstract
The link between the gut microbiome and health has recently garnered considerable interest in its employment for medicinal purposes. Since the early microbiota exhibits more flexibility compared to that of adults, there is a considerable possibility that altering it will have significant consequences on human development. Like genetics, the human microbiota can be passed from mother to child. This provides information on early microbiota acquisition, future development, and prospective chances for intervention. The succession and acquisition of early-life microbiota, modifications of the maternal microbiota during pregnancy, delivery, and infancy, and new efforts to understand maternal-infant microbiota transmission are discussed in this article. We also examine the shaping of mother-to-infant microbial transmission, and we then explore possible paths for future research to advance our knowledge in this area.
Collapse
Affiliation(s)
- Eman A Mady
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan; Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya,13736, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and industrial pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hussein M El-Husseiny
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
241
|
Slack E, Bokulich NA. Microbiomes in Neverland. Cell Host Microbe 2023; 31:461-463. [PMID: 37054668 DOI: 10.1016/j.chom.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Differentiating the effects of infant microbiota, developmental, and nutritional changes on immunological maturation during weaning is an ongoing challenge. In this issue of Cell Host & Microbe, Lubin and colleagues report a gnotobiotic mouse model that maintains neonatal-like microbiome composition into adulthood to help answer burning questions in this field.
Collapse
Affiliation(s)
- Emma Slack
- Laboratory for Mucosal Immunology, Institute of Food, Nutrition, and Health, ETH Zürich, Zürich 8092, Switzerland; Botnar Research Center for Child Health, Basel, Switzerland.
| | - Nicholas A Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition, and Health, ETH Zürich, Zürich 8092, Switzerland
| |
Collapse
|
242
|
Sharma P, Singh S, Das K, Mahant S, Das R. Dysbiosis of gut microbiota due to diet, alcohol intake, body mass index, and gastrointestinal diseases in India. Appl Microbiol Biotechnol 2023; 107:2547-2560. [PMID: 36929191 DOI: 10.1007/s00253-023-12470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
The human gut is composed of diverse microflora which is influenced by dietary intake. Body mass index (BMI) and lifestyle patterns also play a vital role in human health to alter gut microbial composition. Our study aims to determine the impact of alcohol intake, BMI, and diet on gut microbiota and its relationship with gastrointestinal disorders. Thirty-nine gastric biopsies were taken from patients with various gastrointestinal (GI) diseases, and all the patient's lifestyle behavior were recorded in a written proforma. 16S rRNA metagenome analysis for V3-V4 regions was used to examine microbial compositions. The richness and diversity of gut microbiota were analyzed by PERMANOVA using the Bray-Curtis dissimilarity index and principal component analysis. The difference in relative abundance was calculated by ANOVA (p < 0.05). Alpha diversity indexes between vegetarians and non-vegetarians showed no significant difference based on BMI, alcohol status, and GI diseases. We found that in overweight vegetarian individuals Faecalibacterium and Rumicococcus might play a role in the control of Helicobacter pylori. Similarly, the increased abundance of Akkermansia muciniphila in non-vegetarian individuals with normal BMI might play a role to decrease the level of harmful bacteria like H. pylori, and Corynebacterium sp. Also, the relative abundance of Corynebacterium sp. among the vegetarians and Streptococcus sp. in the non-vegetarians was increased in alcoholics while H. pylori was increased in non-alcoholics irrespective of diet. There is an increased abundance of Faecalibacterium prausnitzii in vegetarians among all categories; however, we did not find any correlation between disease outcomes. Our study shows that alcohol intake and dietary habits have independent effects on gut microbial composition. The relative abundance of F. prausnitzii was high among vegetarians in all categories. KEY POINTS: • The presence of H. pylori is less among alcoholics. • Good bacteria help to maintain a normal body mass index. • Gut microbiota richness is high in vegetarians and diversity in non-vegetarians.
Collapse
Affiliation(s)
- Prateek Sharma
- Center for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, 201303, UP, India
| | - Sarika Singh
- Center for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, 201303, UP, India
| | - Kunal Das
- Department of Gastroenterology, Yashoda Super Specialty Hospital, Ghaziabad, UP, India.
| | - Shweta Mahant
- Center for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, 201303, UP, India
| | - Rajashree Das
- Center for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, 201303, UP, India.
| |
Collapse
|
243
|
Cowardin CA, Syed S, Iqbal N, Jamil Z, Sadiq K, Iqbal J, Ali SA, Moore SR. Environmental enteric dysfunction: gut and microbiota adaptation in pregnancy and infancy. Nat Rev Gastroenterol Hepatol 2023; 20:223-237. [PMID: 36526906 PMCID: PMC10065936 DOI: 10.1038/s41575-022-00714-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 03/31/2023]
Abstract
Environmental enteric dysfunction (EED) is a subclinical syndrome of intestinal inflammation, malabsorption and barrier disruption that is highly prevalent in low- and middle-income countries in which poverty, food insecurity and frequent exposure to enteric pathogens impair growth, immunity and neurodevelopment in children. In this Review, we discuss advances in our understanding of EED, intestinal adaptation and the gut microbiome over the 'first 1,000 days' of life, spanning pregnancy and early childhood. Data on maternal EED are emerging, and they mirror earlier findings of increased risks for preterm birth and fetal growth restriction in mothers with either active inflammatory bowel disease or coeliac disease. The intense metabolic demands of pregnancy and lactation drive gut adaptation, including dramatic changes in the composition, function and mother-to-child transmission of the gut microbiota. We urgently need to elucidate the mechanisms by which EED undermines these critical processes so that we can improve global strategies to prevent and reverse intergenerational cycles of undernutrition.
Collapse
Affiliation(s)
- Carrie A Cowardin
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
| | - Sana Syed
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zehra Jamil
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sean R Moore
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
244
|
Abstract
PURPOSE OF REVIEW The increasing global prevalence of food allergy indicates that environmental exposures are likely contributing to food allergy development. This review summarizes recent studies on how specific factors within the external exposome may impact the development of food allergy. RECENT FINDINGS There is strong evidence that nonoral exposure to food allergens within the living environment is a risk factor for food sensitization and food allergy. The role of air pollution in food allergy development remains unclear, as cohort studies have not found consistent relationships between air pollutant exposure and food sensitization. Early-life microbial exposures linked to a rural lifestyle are likely protective against food allergy development, possibly through alteration of the infant microbiome. In contrast, factors associated with urbanization and decreased exposure to microbes may contribute to food allergy development. Recent studies on the role of residential greenness in food allergy development suggest either no relationship or a possible increased risk for food allergy. SUMMARY The external exposome comprises a number of exposures that can modify food allergy risk. Improved understanding of how complex environmental exposures interact with genetic factors will be necessary for developing effective interventions aimed at preventing food allergy development in children.
Collapse
Affiliation(s)
- Timothy P. Moran
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
245
|
Dos Santos SJ, Pakzad Z, Albert AYK, Elwood CN, Grabowska K, Links MG, Hutcheon JA, Maan EJ, Manges AR, Dumonceaux TJ, Hodgson ZG, Lyons J, Mitchell-Foster SM, Gantt S, Joseph K, Van Schalkwyk JE, Hill JE, Money DM. Maternal vaginal microbiome composition does not affect development of the infant gut microbiome in early life. Front Cell Infect Microbiol 2023; 13:1144254. [PMID: 37065202 PMCID: PMC10097898 DOI: 10.3389/fcimb.2023.1144254] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/07/2023] [Indexed: 04/01/2023] Open
Abstract
Birth mode has been implicated as a major factor influencing neonatal gut microbiome development, and it has been assumed that lack of exposure to the maternal vaginal microbiome is responsible for gut dysbiosis among caesarean-delivered infants. Consequently, practices to correct dysbiotic gut microbiomes, such as vaginal seeding, have arisen while the effect of the maternal vaginal microbiome on that of the infant gut remains unknown. We conducted a longitudinal, prospective cohort study of 621 Canadian pregnant women and their newborn infants and collected pre-delivery maternal vaginal swabs and infant stool samples at 10-days and 3-months of life. Using cpn60-based amplicon sequencing, we defined vaginal and stool microbiome profiles and evaluated the effect of maternal vaginal microbiome composition and various clinical variables on the development of the infant stool microbiome. Infant stool microbiomes showed significant differences in composition by delivery mode at 10-days postpartum; however, this effect could not be explained by maternal vaginal microbiome composition and was vastly reduced by 3 months. Vaginal microbiome clusters were distributed across infant stool clusters in proportion to their frequency in the overall maternal population, indicating independence of the two communities. Intrapartum antibiotic administration was identified as a confounder of infant stool microbiome differences and was associated with lower abundances of Escherichia coli, Bacteroides vulgatus, Bifidobacterium longum and Parabacteroides distasonis. Our findings demonstrate that maternal vaginal microbiome composition at delivery does not affect infant stool microbiome composition and development, suggesting that practices to amend infant stool microbiome composition focus factors other than maternal vaginal microbes.
Collapse
Affiliation(s)
- Scott J. Dos Santos
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zahra Pakzad
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
- Women’s Health Research Institute, B.C. Women's Hopsital, Vancouver, BC, Canada
| | | | - Chelsea N. Elwood
- Women’s Health Research Institute, B.C. Women's Hopsital, Vancouver, BC, Canada
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kirsten Grabowska
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Matthew G. Links
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Computer Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jennifer A. Hutcheon
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Evelyn J. Maan
- Women’s Health Research Institute, B.C. Women's Hopsital, Vancouver, BC, Canada
| | - Amee R. Manges
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | | | - Zoë G. Hodgson
- Midwifery Program, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Janet Lyons
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sheona M. Mitchell-Foster
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Soren Gantt
- Centre de Recherche du CHU Sainte-Justine, Montréal, QC, Canada
| | - K.S. Joseph
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Julie E. Van Schalkwyk
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Janet E. Hill
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Deborah M. Money, ; Janet E. Hill,
| | - Deborah M. Money
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
- Women’s Health Research Institute, B.C. Women's Hopsital, Vancouver, BC, Canada
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Deborah M. Money, ; Janet E. Hill,
| |
Collapse
|
246
|
Shaikh SD, Sun N, Canakis A, Park WY, Weber HC. Irritable Bowel Syndrome and the Gut Microbiome: A Comprehensive Review. J Clin Med 2023; 12:jcm12072558. [PMID: 37048642 PMCID: PMC10095554 DOI: 10.3390/jcm12072558] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Irritable Bowel Syndrome (IBS) is a functional disorder of the gastrointestinal tract characterized by abdominal pain and altered bowel habits. It has a prevalence of 10 to 25% in the United States and has a high disease burden, as evidenced by reduced quality of life, decreased work productivity and increased healthcare utilization and costs. IBS has been associated with several intra-intestinal and extra-intestinal conditions, including psychiatric comorbidities. Although the pathophysiology of IBS has not been fully elucidated, it involves dysregulation of communication between the brain and gut (brain–gut axis) which is associated with alterations in intestinal motility, gut permeability, visceral hypersensitivity and gut microbiota composition. The purpose of this article is to review the role the gut microbiota plays in the pathophysiology of IBS, understand factors that affect the gut microbiome and explore the microbiome as a target of treatment.
Collapse
|
247
|
Pieri M, Nicolaidou V, Papaneophytou C. Special Issue: The Impact of Early Life Nutrition on Gut Maturation and Later Life Gut Health. Nutrients 2023; 15:nu15061498. [PMID: 36986228 PMCID: PMC10058133 DOI: 10.3390/nu15061498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Nutrition during early life plays a crucial role in determining a child's long-term health [...].
Collapse
Affiliation(s)
- Myrtani Pieri
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| | - Vicky Nicolaidou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| |
Collapse
|
248
|
Stiernborg M, Debelius JW, Yang LL, Skott E, Millischer V, Giacobini M, Melas PA, Boulund F, Lavebratt C. Bacterial gut microbiome differences in adults with ADHD and in children with ADHD on psychostimulant medication. Brain Behav Immun 2023; 110:310-321. [PMID: 36940753 DOI: 10.1016/j.bbi.2023.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/11/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Recent evidence suggests that there is a link between neurodevelopmental disorders, such as attention deficit hyperactivity disorder (ADHD), and the gut microbiome. However, most studies to date have had low sample sizes, have not investigated the impact of psychostimulant medication, and have not adjusted for potential confounders, including body mass index, stool consistency and diet. To this end, we conducted the largest, to our knowledge, fecal shotgun metagenomic sequencing study in ADHD, with 147 well-characterized adult and child patients. For a subset of individuals, plasma levels of inflammatory markers and short-chain fatty acids were also measured. In adult ADHD patients (n=84), compared to controls (n=52), we found a significant difference in beta diversity both regarding bacterial strains (taxonomic) and bacterial genes (functional). In children with ADHD (n=63), we found that those on psychostimulant medication (n=33 on medication vs. n=30 not on medication) had (i) significantly different taxonomic beta diversity, (ii) lower functional and taxonomic evenness, (iii) lower abundance of the strain Bacteroides stercoris CL09T03C01 and bacterial genes encoding an enzyme in vitamin B12 synthesis, and (iv) higher plasma levels of vascular inflammatory markers sICAM-1 and sVCAM-1. Our study continues to support a role for the gut microbiome in neurodevelopmental disorders and provides additional insights into the effects of psychostimulant medication. However, additional studies are needed to replicate these findings and examine causal relationships with the disorder.
Collapse
Affiliation(s)
- Miranda Stiernborg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - J W Debelius
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; The Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Liu L Yang
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden; Department of Neurology, Huazhong University of Science and Technology, Tongji Medical College, Union Hospital, Wuhan, China
| | - Elin Skott
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden; PRIMA Child and Adult Psychiatry, Stockholm, Sweden
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - MaiBritt Giacobini
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; PRIMA Child and Adult Psychiatry, Stockholm, Sweden
| | - Philippe A Melas
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden; Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm, Sweden
| | - Fredrik Boulund
- The Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden.
| |
Collapse
|
249
|
Wang Y, Zhu J, Zou N, Zhang L, Wang Y, Zhang M, Wang C, Yang L. Pathogenesis from the microbial-gut-brain axis in white matter injury in preterm infants: A review. Front Integr Neurosci 2023; 17:1051689. [PMID: 37006416 PMCID: PMC10060642 DOI: 10.3389/fnint.2023.1051689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
White matter injury (WMI) in premature infants is a unique form of brain injury and a common cause of chronic nervous system conditions such as cerebral palsy and neurobehavioral disorders. Very preterm infants who survive are at high risk of WMI. With developing research regarding the pathogenesis of premature WMI, the role of gut microbiota has attracted increasing attention in this field. As premature infants are a special group, early microbial colonization of the microbiome can affect brain development, and microbiome optimization can improve outcomes regarding nervous system development. As an important communication medium between the gut and the nervous system, intestinal microbes form a microbial-gut-brain axis. This axis affects the occurrence of WMI in premature infants via the metabolites produced by intestinal microorganisms, while also regulating cytokines and mediating oxidative stress. At the same time, deficiencies in the microbiota and their metabolites may exacerbate WMI in premature infants. This confers promise for probiotics and prebiotics as treatments for improving neurodevelopmental outcomes. Therefore, this review attempted to elucidate the potential mechanisms behind the communication of gut bacteria and the immature brain through the gut-brain axis, so as to provide a reference for further prevention and treatment of premature WMI.
Collapse
|
250
|
Abbes S, Baldi S, Sellami H, Amedei A, Keskes L. Molecular methods for colorectal cancer screening: Progress with next-generation sequencing evolution. World J Gastrointest Oncol 2023; 15:425-442. [PMID: 37009313 PMCID: PMC10052664 DOI: 10.4251/wjgo.v15.i3.425] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 02/14/2023] [Indexed: 03/14/2023] Open
Abstract
Currently, colorectal cancer (CRC) represents the third most common malignancy and the second most deadly cancer worldwide, with a higher incidence in developed countries. Like other solid tumors, CRC is a heterogeneous genomic disease in which various alterations, such as point mutations, genomic rearrangements, gene fusions or chromosomal copy number alterations, can contribute to the disease development. However, because of its orderly natural history, easily accessible onset location and high lifetime incidence, CRC is ideally suited for preventive intervention, but the many screening efforts of the last decades have been compromised by performance limitations and low penetrance of the standard screening tools. The advent of next-generation sequencing (NGS) has both facilitated the identification of previously unrecognized CRC features such as its relationship with gut microbial pathogens and revolutionized the speed and throughput of cataloguing CRC-related genomic alterations. Hence, in this review, we summarized the several diagnostic tools used for CRC screening in the past and the present, focusing on recent NGS approaches and their revolutionary role in the identification of novel genomic CRC characteristics, the advancement of understanding the CRC carcinogenesis and the screening of clinically actionable targets for personalized medicine.
Collapse
Affiliation(s)
- Salma Abbes
- Laboratory of Parasitic and Fungal Molecular Biology, University of Sfax, Sfax 3029, Tunisia
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Hayet Sellami
- Drosophila Research Unit-Parasitology and Mycologie Laboratory, University of Sfax, Sfax 3029, Tunisia
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- SOD of Interdisciplinary Internal Medicine, Careggi University Hospital, Florence 50134, Italy
| | - Leila Keskes
- Laboratory of Human Molecular Genetic, University of Sfax, Sfax 3029, Tunisia
| |
Collapse
|