1
|
Miller C, Luu K, Mikami B, Riel J, Qin Y, Khadka V, Lee MJ. Temporal Investigation of the Maternal Origins of Fetal Gut Microbiota. Microorganisms 2024; 12:1865. [PMID: 39338539 PMCID: PMC11434507 DOI: 10.3390/microorganisms12091865] [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: 07/26/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
In utero colonization or deposition of beneficial microorganisms and their by-products likely occurs through various mechanisms, such as hematogenous spread or ascension from the reproductive tract. With high-throughput sequencing techniques, the identification of microbial components in first-pass neonatal meconium has been achieved. While these components are low-biomass and often not abundant enough to culture, the presence of microbial DNA signatures may promote fetal immune tolerance or epigenetic regulation prior to birth. The aim of this study was to investigate the maternal source of the neonatal first-pass meconium microbiome. Maternal vaginal and anal samples collected from twenty-one maternal-infant dyad pairs were compared via principal component analysis to first-pass neonatal meconium microbial compositions. Results demonstrated the greatest degree of similarity between the maternal gut microbiome in the second and third trimesters and vaginal microbiome samples across pregnancy, suggesting that the maternal gut microbiota may translocate to the fetal gut during pregnancy. This study sheds light on the origin and timing of the potential transfer of maternal microbial species to offspring in utero.
Collapse
Affiliation(s)
- Corrie Miller
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal Fetal Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Kayti Luu
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal Fetal Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Brandi Mikami
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal Fetal Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Jonathan Riel
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal Fetal Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Yujia Qin
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Vedbar Khadka
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Men-Jean Lee
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal Fetal Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
2
|
Sasaki T, Kawamura M, Okuno C, Lau K, Riel J, Lee MJ, Miller C. Impact of Maternal Mediterranean-Type Diet Adherence on Microbiota Composition and Epigenetic Programming of Offspring. Nutrients 2023; 16:47. [PMID: 38201877 PMCID: PMC10780434 DOI: 10.3390/nu16010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Understanding how maternal diet affects in utero neonatal gut microbiota and epigenetic regulation may provide insight into disease origins and long-term health. The impact of Mediterranean diet pattern adherence (MDA) on fetal gut microbiome and epigenetic regulation was assessed in 33 pregnant women. Participants completed a validated food frequency questionnaire in each trimester of pregnancy; the alternate Mediterranean diet (aMED) score was applied. Umbilical cord blood, placental tissue, and neonatal meconium were collected from offspring. DNA methylation patterns were probed using the Illumnia EPICarray Methylation Chip in parturients with high versus low MDA. Meconium microbial abundance in the first 24 h after birth was identified using 16s rRNA sequencing and compared among neonates born to mothers with high and low aMED scores. Twenty-one mothers were classified as low MDA and 12 as high MDA. Pasteurellaceae and Bacteroidaceae trended towards greater abundance in the high-MDA group, as well as other short-chain fatty acid-producing species. Several differentially methylated regions varied between groups and overlapped gene regions including NCK2, SNED1, MTERF4, TNXB, HLA-DPB, BAG6, and LMO3. We identified a beneficial effect of adherence to a Mediterranean diet on fetal in utero development. This highlights the importance of dietary counseling for mothers and can be used as a guide for future studies of meconium and immuno-epigenetic modulation.
Collapse
Affiliation(s)
- Tamlyn Sasaki
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Megan Kawamura
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Chirstyn Okuno
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Kayleen Lau
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Jonathan Riel
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96826, USA
| | - Men-Jean Lee
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96826, USA
| | - Corrie Miller
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96826, USA
| |
Collapse
|
3
|
Liu D, Wang X, Zhang H. Efficacy and safety of gastrointestinal microbiome supplementation for allergic rhinitis: A systematic review and meta-analysis with trial sequential analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154948. [PMID: 37418839 DOI: 10.1016/j.phymed.2023.154948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/07/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) is a non-infective chronic inflammatory disease of nasal mucosa. PURPOSE To evaluate the efficacy and safety of gastrointestinal microbiome supplementation (GMS) for patients with allergic rhinitis (AR), concerning improvement on symptoms and signs, laboratory outcomes, quality of life, and medication scores. METHODS Five English databases were searched up to Dec 12th, 2022. Probiotics, prebiotics, and synbiotics were main therapies or adjuvants in experimental groups. Systematic reviews and meta-analyses were conducted based on the Cochrane systematic review method by using RevMan 5.4 Software, with meta-influence analyses, subgroup-analyses, meta-regression, and publication bias performed for exploration of heterogeneity by Stata V.14. Trial sequential analyses were performed by TSA 0.9, and quality of the results was accessed through the GRADE-pro GDT. RESULTS Finally, extracted from 53 articles, 65 RCTs involving 3,634 participants with sound worldwide representativeness were included. Primary results showed better improvement in GMS groups on TNSS (WMD=1.05, P for WMD=0.004, 95%CI:0.34 to 1.76), overall nasal condition (WMD=1.25, P for WMD<0.001, 95%CI:0.90 to 1.61), overall quality of life (WMD=6.16, P for WMD<0.001, 95%CI:4.92 to 7.40) and medication score (WMD=0.42, P for WMD=0.42, 95%CI:-0.06 to 0.90).However, GMS groups were inferior than the controls concerning reduction on serum total IgE (WMD=-1.81) and ratios of serum Th1/Th2 (WMD=-1.06). Meta-regressions suggested significant (p<0.05) variations of the effects in some comparisons. In addition, results of sub-group analyses firstly revealed potential influence between final results and the variables above. Instantly after intervention, the GRADE levels of evidence were sound, including "High ⨁⨁⨁⨁" in 10, "Moderate ⨁⨁⨁◯" in 33, and "Low ⨁⨁◯◯" in nine comparisons. However, overall certainties decreased obviously during follow-ups. CONCLUSION Overall, our pooled results firstly revealed that GMS yielded acceptable benefits for patients with AR compared with controls with sound certainties, after balancing the benefits and harms.
Collapse
Affiliation(s)
- Dongliang Liu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, PR China
| | - Xilu Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, PR China
| | - Hang Zhang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang 110004, PR China.
| |
Collapse
|
4
|
Metagenomic profiles of the early life microbiome of Indonesian inpatient neonates and their influence on clinical characteristics. Sci Rep 2022; 12:9413. [PMID: 35672441 PMCID: PMC9174262 DOI: 10.1038/s41598-022-13496-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/25/2022] [Indexed: 11/21/2022] Open
Abstract
Determining the initial normal neonatal gut microbiome is challenging. The debate regarding the sterile fetal environment is still ongoing. Therefore, studying and comparing normal and dysbiotic microbiomes requires the elucidation of both the fetal and infant microbiomes. Factors influencing the normal microbiome also include regional and genetic factors specific to different countries. Determining the normal microbiome population in our center and their association with the clinical conditions of infants is helpful as a tool for both the prevention and treatment of related diseases during neonatal care. Here, we employed metagenomic sequencing to characterize meconium and the subsequent early-life gut microbiome of preterm neonates in Jakarta, Indonesia. Microbiome diversity and complexity was higher in the meconium and on day 4 than on day 7. At the genus level, the most abundant genus overall was unidentified Enterobacteriaceae, with meconium samples dominated by Ureaplasma, day 4 fecal samples dominated by Staphylococcus, and day 7 samples dominated by Clostridiales, while at the phylum level the most abundant was Proteobacteria and Firmicutes. Perinatal factors of PROM and mother’s diet influenced the meconium microbiome, while day 4 and day 7 microbiome was associated with bacteremia and early administration of antibiotics. One of our sample sets was derived from triplets, and they had varying diversity despite being triplets. These data are valuable for understanding the formation of a healthy microbiome specific to neonates and devising a strategy to improve both the gut health and related clinical outcomes of the neonate.
Collapse
|
5
|
Farinella R, Rizzato C, Bottai D, Bedini A, Gemignani F, Landi S, Peduzzi G, Rosati S, Lupetti A, Cuttano A, Moscuzza F, Tuoni C, Filippi L, Ciantelli M, Tavanti A, Campa D. Maternal anthropometric variables and clinical factors shape neonatal microbiome. Sci Rep 2022; 12:2875. [PMID: 35190600 PMCID: PMC8861021 DOI: 10.1038/s41598-022-06792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/30/2021] [Indexed: 11/09/2022] Open
Abstract
AbstractRecent studies indicate the existence of a complex microbiome in the meconium of newborns that plays a key role in regulating many host health-related conditions. However, a high variability between studies has been observed so far. In the present study, the meconium microbiome composition and the predicted microbial metabolic pathways were analysed in a consecutive cohort of 96 full-term newborns. The effect of maternal epidemiological variables on meconium diversity was analysed using regression analysis and PERMANOVA. Meconium microbiome composition mainly included Proteobacteria (30.95%), Bacteroidetes (23.17%) and Firmicutes (17.13%), while for predicted metabolic pathways, the most abundant genes belonged to the class “metabolism”. We observed a significant effect of maternal Rh factor on Shannon and Inverse Simpson indexes (p = 0.045 and p = 0.049 respectively) and a significant effect of delivery mode and maternal antibiotic exposure on Jaccard and Bray–Curtis dissimilarities (p = 0.001 and 0.002 respectively), while gestational age was associated with observed richness and Shannon indexes (p = 0.018 and 0.037 respectively), and Jaccard and Bray–Curtis dissimilarities (p = 0.014 and 0.013 respectively). The association involving maternal Rh phenotype suggests a role for host genetics in shaping meconium microbiome prior to the exposition to the most well-known environmental variables, which will influence microbiome maturation in the newborn.
Collapse
|
6
|
Poier G, Terler G, Klevenhusen F, Sharma S, Zebeli Q. Replacing concentrates with a high-quality hay in the starter feed of dairy calves: II. Effects on the development of chewing and gut fermentation, and selected systemic health variables. J Dairy Sci 2022; 105:3113-3128. [DOI: 10.3168/jds.2021-21346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/08/2021] [Indexed: 11/19/2022]
|
7
|
Abstract
Meconium constitutes infants' first bowel movements postnatally. The consistency and microbial load of meconium are different from infant and adult stool. While recent evidence suggests that meconium is sterile in utero, rapid colonization occurs after birth. The meconium microbiome has been associated with negative health outcomes, but its composition is not well described, especially in preterm infants. Here, we characterized the meconium microbiomes from 330 very preterm infants (gestational ages 28 to 32 weeks) from 15 hospitals in Germany and in fecal samples from a subset of their mothers (N = 217). Microbiome profiles were compiled using 16S rRNA gene sequencing with negative and positive controls. The meconium microbiome was dominated by Bifidobacterium, Staphylococcus, and Enterococcus spp. and was associated with gestational age at birth and age at sample collection. Bifidobacterial abundance was negatively correlated with potentially pathogenic genera. The amount of bacterial DNA in meconium samples varied greatly across samples and was associated with the time since birth but not with gestational age or hospital site. In samples with low bacterial load, human mitochondrial sequences were highly amplified using commonly used, bacterial-targeted 16S rRNA primers. Only half of the meconium samples contained sufficient bacterial material to study the microbiome using a standard approach. To facilitate future meconium studies, we present a five-level scoring system (“MecBac”) that predicts the success of 16S rRNA bacterial sequencing for meconium samples. These findings provide a foundational characterization of an understudied portion of the human microbiome and will aid the design of future meconium microbiome studies. IMPORTANCE Meconium is present in the intestines of infants before and after birth and constitutes their first bowel movements postnatally. The consistency, composition and microbial load of meconium is largely different from infant and adult stool. While recent evidence suggests that meconium is sterile in utero, rapid colonization occurs after birth. The meconium microbiome has been associated with short-term and long-term negative health outcomes, but its composition is not yet well described, especially in preterm infants. We provide a characterization of the microbiome structure and composition of infant meconium and maternal feces from a large study cohort and propose a method to evaluate meconium samples for bacterial sequencing suitability. These findings provide a foundational characterization of an understudied portion of the human microbiome and will aid the design of future meconium microbiome studies.
Collapse
|
8
|
Abstract
The maternal microbiome has emerged as an important area of investigation. While birth is a critical timepoint for initial colonization of the newborn, the fetus resides in the womb surrounded by multiple unique colonized niches. The maternal microbiome has recently been shown to be associated with several morbidities in offspring. Understanding the multiple bacterial niches within the pregnant woman and how they interact with the fetus in-utero can lead to novel therapies to improve the health of offspring. In this review, we provide an overview of the available literature on normal bacterial colonization within the individual niches of the pregnant woman and the known associations with outcomes in offspring, including a discussion of the controversy of in-utero colonization.
Collapse
Affiliation(s)
- Julie Mirpuri
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Josef Neu
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
9
|
Padmanabhan V, Song W, Puttabyatappa M. Praegnatio Perturbatio-Impact of Endocrine-Disrupting Chemicals. Endocr Rev 2021; 42:295-353. [PMID: 33388776 PMCID: PMC8152448 DOI: 10.1210/endrev/bnaa035] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Indexed: 02/07/2023]
Abstract
The burden of adverse pregnancy outcomes such as preterm birth and low birth weight is considerable across the world. Several risk factors for adverse pregnancy outcomes have been identified. One risk factor for adverse pregnancy outcomes receiving considerable attention in recent years is gestational exposure to endocrine-disrupting chemicals (EDCs). Humans are exposed to a multitude of environmental chemicals with known endocrine-disrupting properties, and evidence suggests exposure to these EDCs have the potential to disrupt the maternal-fetal environment culminating in adverse pregnancy and birth outcomes. This review addresses the impact of maternal and fetal exposure to environmental EDCs of natural and man-made chemicals in disrupting the maternal-fetal milieu in human leading to adverse pregnancy and birth outcomes-a risk factor for adult-onset noncommunicable diseases, the role lifestyle and environmental factors play in mitigating or amplifying the effects of EDCs, the underlying mechanisms and mediators involved, and the research directions on which to focus future investigations to help alleviate the adverse effects of EDC exposure.
Collapse
Affiliation(s)
| | - Wenhui Song
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
10
|
Li YF, Gong XL, Chen SX, Wang K, Jiang YH. Deviations in the gut microbiota of neonates affected by maternal group B Streptococcus colonization. BMC Microbiol 2021; 21:140. [PMID: 33952201 PMCID: PMC8097833 DOI: 10.1186/s12866-021-02204-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/23/2021] [Indexed: 01/30/2023] Open
Abstract
Background Group B Streptococcus (GBS) infection is the leading cause of septicemia, meningitis, and pneumonia in neonates. Aberrant gut colonization in early life may predispose children to various diseases in adulthood. However, the associations between gut microbial changes and GBS colonization is still unclear. Results The composition and diversity of meconium microbiota in GBS group were similar to that of healthy controls. However, we identified several specific taxa that were differentially abundant between the two groups (linear discriminant analysis (LDA) effect size (LEfSe): p < 0.05, LDA > 2.0). Particularly, the relative abundance of Lactobacillus paracasei was significantly reduced, indicating a role in GBS colonization. Conclusions Our study presented a series of bacterial species colonized by GBS, thus providing novel evidence in support of initial intestinal microbiota dysbiosis in the neonates with mother’s GBS colonization. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02204-3.
Collapse
Affiliation(s)
- Yue-Feng Li
- Department of Pediatrics, Shenzhen Luohu Maternity and Child Health Hospital, Shenzhen, 518019, China
| | - Xue-Lei Gong
- Department of Pediatrics, Shenzhen Luohu Maternity and Child Health Hospital, Shenzhen, 518019, China
| | - Su-Xiang Chen
- Department of Pediatrics, Shenzhen Luohu Maternity and Child Health Hospital, Shenzhen, 518019, China
| | - Kejian Wang
- The Third Affiliated Hospital of Shandong First Medical University Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, 250031, China.
| | - Yan-Hua Jiang
- Departments of Obstetrics and Gynecology, Shenzhen Luohu Maternity and Child Health Hospital, No.2013, Taibai Road, Luohu District, Shenzhen, 518019, China.
| |
Collapse
|
11
|
Contribution of Infectious Agents to the Development of Celiac Disease. Microorganisms 2021; 9:microorganisms9030547. [PMID: 33800833 PMCID: PMC8001938 DOI: 10.3390/microorganisms9030547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
The ingestion of wheat gliadin (alcohol-soluble proteins, an integral part of wheat gluten) and related proteins induce, in genetically predisposed individuals, celiac disease (CD), which is characterized by immune-mediated impairment of the small intestinal mucosa. The lifelong omission of gluten and related grain proteins, i.e., a gluten-free diet (GFD), is at present the only therapy for CD. Although a GFD usually reduces CD symptoms, it does not entirely restore the small intestinal mucosa to a fully healthy state. Recently, the participation of microbial components in pathogenetic mechanisms of celiac disease was suggested. The present review provides information on infectious diseases associated with CD and the putative role of infections in CD development. Moreover, the involvement of the microbiota as a factor contributing to pathological changes in the intestine is discussed. Attention is paid to the mechanisms by which microbes and their components affect mucosal immunity, including tolerance to food antigens. Modulation of microbiota composition and function and the potential beneficial effects of probiotics in celiac disease are discussed.
Collapse
|
12
|
Olshan KL, Leonard MM, Serena G, Zomorrodi AR, Fasano A. Gut microbiota in Celiac Disease: microbes, metabolites, pathways and therapeutics. Expert Rev Clin Immunol 2020; 16:1075-1092. [PMID: 33103934 DOI: 10.1080/1744666x.2021.1840354] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Current evidence supports a vital role of the microbiota on health outcomes, with alterations in an otherwise healthy balance linked to chronic medical conditions like celiac disease (CD). Recent advances in microbiome analysis allow for unparalleled profiling of the microbes and metabolites. With the growing volume of data available, trends are emerging that support a role for the gut microbiota in CD pathogenesis. AREAS COVERED In this article, the authors review the relationship between factors such as genes and antibiotic exposure on CD onset and the intestinal microbiota. The authors also review other microbiota within the human body (like the oropharynx) that may play a role in CD pathogenesis. Finally, the authors discuss implications for disease modification and the ultimate goal of prevention. The authors reviewed literature from PubMed, EMBASE, and Web of Science. EXPERT OPINION CD serves as a unique opportunity to explore the role of the intestinal microbiota on the development of chronic autoimmune disease. While research to date provides a solid foundation, most studies have been case-control and thus do not have capacity to explore the mechanistic role of the microbiota in CD onset. Further longitudinal studies and integrated multi-omics are necessary for investigating CD pathogenesis.
Collapse
Affiliation(s)
- Katherine L Olshan
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Ali R Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Celiac Research Program, Harvard Medical School , Boston, MA, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School , Boston, MA, USA.,European Biomedical Research Institute of Salerno (EBRIS) , Salerno, Italy
| |
Collapse
|
13
|
Selma-Royo M, Calatayud Arroyo M, García-Mantrana I, Parra-Llorca A, Escuriet R, Martínez-Costa C, Collado MC. Perinatal environment shapes microbiota colonization and infant growth: impact on host response and intestinal function. MICROBIOME 2020; 8:167. [PMID: 33228771 PMCID: PMC7685601 DOI: 10.1186/s40168-020-00940-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/14/2020] [Indexed: 05/11/2023]
Abstract
BACKGROUND Early microbial colonization triggers processes that result in intestinal maturation and immune priming. Perinatal factors, especially those associated with birth, including both mode and place of delivery are critical to shaping the infant gut microbiota with potential health consequences. METHODS Gut microbiota profile of 180 healthy infants (n = 23 born at home and n = 157 born in hospital, 41.7% via cesarean section [CS]) was analyzed by 16S rRNA gene sequencing at birth, 7 days, and 1 month of life. Breastfeeding habits and infant clinical data, including length, weight, and antibiotic exposure, were collected up to 18 months of life. Long-term personalized in vitro models of the intestinal epithelium and innate immune system were used to assess the link between gut microbiota composition, intestinal function, and immune response. RESULTS Microbiota profiles were shaped by the place and mode of delivery, and they had a distinct biological impact on the immune response and intestinal function in epithelial/immune cell models. Bacteroidetes and Bifidobacterium genus were decreased in C-section infants, who showed higher z-scores BMI and W/L during the first 18 months of life. Intestinal simulated epithelium had a stronger epithelial barrier function and intestinal maturation, alongside a higher immunological response (TLR4 route activation and pro-inflammatory cytokine release), when exposed to home-birth fecal supernatants, compared with CS. Distinct host response could be associated with different microbiota profiles. CONCLUSIONS Mode and place of birth influence the neonatal gut microbiota, likely shaping its interplay with the host through the maturation of the intestinal epithelium, regulation of the intestinal epithelial barrier, and control of the innate immune system during early life, which can affect the phenotypic responses linked to metabolic processes in infants. TRIAL REGISTRATION NCT03552939 . Video Abstract.
Collapse
Affiliation(s)
- M Selma-Royo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - M Calatayud Arroyo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - I García-Mantrana
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - A Parra-Llorca
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain
| | - R Escuriet
- Gerencia de Procesos Integrales de Salud. Area Asistencial, Servicio Catalan de la Salud. Generalitat de Catalunya, Centre for Research in Health and Economics, Universidad Pompeu Fabra, Barcelona, Spain
| | - C Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, 46010, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, 46010, Valencia, Spain
| | - M C Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain.
| |
Collapse
|
14
|
Morais J, Marques C, Teixeira D, Durão C, Faria A, Brito S, Cardoso M, Macedo I, Pereira E, Tomé T, Calhau C. Extremely preterm neonates have more Lactobacillus in meconium than very preterm neonates - the in utero microbial colonization hypothesis. Gut Microbes 2020; 12:1785804. [PMID: 32658601 PMCID: PMC7524394 DOI: 10.1080/19490976.2020.1785804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Growing evidence suggests that maternal microbiota can influence the neonates' gut colonization. However, the mechanisms of vertical bacterial transmission remain poorly defined. We believed that the first colonizers of the newborn come from the mother's gut and vagina during pregnancy and that this is independent of the mode of delivery. We conducted an observational longitudinal study to evaluate the link between the maternal gut microbiota and the meconium's microbiota in extremely and very preterm neonates. Bacterial DNA was extracted from samples and specific bacterial groups were quantified by RT-PCR. In this cohort of 117 preterm neonates, we detected bacterial DNA in 88% of meconium samples. Meconium microbiota of neonates born after 28 gestational weeks (very preterm neonates) showed stronger correlations with their mothers' fecal microbiota. However, neonates born before 28 gestational weeks (extremely preterm neonates) had more Lactobacillus - genus that dominated the vaginal microbiota - than very preterm neonates, regardless of the mode of delivery. Collectively, these data support the hypothesis that maternal bacteria from the gut and vagina can play a role in shaping neonates' gut microbiota and that mother-to-infant bacterial transmission is a controlled and time-specific process. ClinicalTrials.gov Identifier: NCT03663556.
Collapse
Affiliation(s)
- Juliana Morais
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,CINTESIS, Center for Health Technology Services Research, Porto, Portugal
| | - Cláudia Marques
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,CINTESIS, Center for Health Technology Services Research, Porto, Portugal
| | - Diana Teixeira
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,Comprehensive Health Research Centre, Universidade NOVA de Lisboa, Lisboa, Portugal,Unidade Universitária Lifestyle Medicine José de Mello Saúde by NOVA Medical School, Lisboa, Portugal
| | - Catarina Durão
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,EPIUnit - Institute of Public Health, Universidade do Porto, Porto, Portugal
| | - Ana Faria
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,CINTESIS, Center for Health Technology Services Research, Porto, Portugal,Comprehensive Health Research Centre, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Sara Brito
- Neonatal Intensive Care Unit, Liutrition and Dietetics Unit, Centro Hospitalar Universitário de Lisboa Central, Maternidade Dr. Alfredo da Costa, Cesboa, Portugal
| | - Manuela Cardoso
- Nutrition and Dietetics Unit, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Israel Macedo
- Neonatal Intensive Care Unit, Liutrition and Dietetics Unit, Centro Hospitalar Universitário de Lisboa Central, Maternidade Dr. Alfredo da Costa, Cesboa, Portugal
| | - Esmeralda Pereira
- Neonatology Department, Maternidade Dr. Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Teresa Tomé
- Neonatal Intensive Care Unit, Liutrition and Dietetics Unit, Centro Hospitalar Universitário de Lisboa Central, Maternidade Dr. Alfredo da Costa, Cesboa, Portugal
| | - Conceição Calhau
- Nutrition and Metabolism, Faculdade de Ciências Médicas
- NOVA Medical Schoo
- , Universidade NOVA de Lisboa, Lisboa, Portugal,CINTESIS, Center for Health Technology Services Research, Porto, Portugal,Unidade Universitária Lifestyle Medicine José de Mello Saúde by NOVA Medical School, Lisboa, Portugal,CONTACT Conceição Calhau, Faculdade de Ciências Médicas
- NOVA Medical School, Universidade NOVA de Lisboa, Lisboa1169-056, Portugal
| |
Collapse
|
15
|
Fernández-Pérez S, Pérez-Andrés J, Gutiérrez S, Navasa N, Martínez-Blanco H, Ferrero MÁ, Vivas S, Vaquero L, Iglesias C, Casqueiro J, Rodríguez-Aparicio LB. The Human Digestive Tract Is Capable of Degrading Gluten from Birth. Int J Mol Sci 2020; 21:ijms21207696. [PMID: 33080976 PMCID: PMC7589136 DOI: 10.3390/ijms21207696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
The human gastrointestinal system has the capacity to metabolize dietary gluten. The capacity to degrade gliadin-derived peptide is present in humans from birth and increases during the first stages of life (up to 6–12 months of age). Fecal samples from 151 new-born and adult non-celiac disease (NCD) volunteers were collected, and glutenase and glianidase activities were evaluated. The capacity of total fecal proteins to metabolize 33-mer, 19-mer, and 13-mer gliadin peptides was also evaluated by high-performance liquid chromatography (HPLC). Feces from new-borns (meconium) showed glutenase and gliadinase activities, and peptidase activity against all three gliadin peptides. Maximal gluten degradative activity was observed in fecal samples from the youngest volunteers (0–12 months old). After the age of nine months, the gluten digestive capacity of gastrointestinal tract decreases and, from ±8 years old, individuals lose the ability to completely degrade toxic peptides. The gastrointestinal proteases involved in gluten digestion: elastase 2A, elastase 3B, and carboxipeptidase A1 are present from earlier stages of life. The human digestive tract contains the proteins capable of metabolizing gluten from birth, even before starting gluten intake. Humans are born with the ability to digest gluten and to completely degrade the potentially toxic gliadin-derived peptides (33-, 19-, and 13-mer).
Collapse
Affiliation(s)
- Silvia Fernández-Pérez
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Jenifer Pérez-Andrés
- Área de Microbiología, Departamento de Biología Molecular, Facultad de Ciencias Biológicas y Ambientales, Universidad de León, 24071 León, Spain; (J.P.-A.); (J.C.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Sergio Gutiérrez
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Nicolás Navasa
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Honorina Martínez-Blanco
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Miguel Ángel Ferrero
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Santiago Vivas
- Servicio de Gastroenterología, Hospital Universitario de León, 24008 Léon, Spain; (S.V.); (L.V.); (C.I.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
| | - Luis Vaquero
- Servicio de Gastroenterología, Hospital Universitario de León, 24008 Léon, Spain; (S.V.); (L.V.); (C.I.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain
| | - Cristina Iglesias
- Servicio de Gastroenterología, Hospital Universitario de León, 24008 Léon, Spain; (S.V.); (L.V.); (C.I.)
- Servicio de Pediatría, Hospital Universitario de León, 24008 Léon, Spain
| | - Javier Casqueiro
- Área de Microbiología, Departamento de Biología Molecular, Facultad de Ciencias Biológicas y Ambientales, Universidad de León, 24071 León, Spain; (J.P.-A.); (J.C.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Leandro B. Rodríguez-Aparicio
- Área de Bioquímica y Biología Molecular, Departamento de Biología Molecular, Facultad de Veterinaria, Universidad de León, 24071 León, Spain; (S.F.-P.); (S.G.); (N.N.); (H.M.-B.); (M.Á.F.)
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
- Correspondence: ; Tel.: +34-987-291227
| |
Collapse
|
16
|
Yang J, Yao S, Cheng K, Xu L, Hou L, Wei Y, Feng H, Yu X, Zhang X, Tong X, Li Z, Zhao Y. Comparison of Meconium Microbiome in Dizygotic and Monozygotic Twins Born by Caesarean Section (CS). Front Microbiol 2020; 11:1139. [PMID: 32582089 PMCID: PMC7283445 DOI: 10.3389/fmicb.2020.01139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/05/2020] [Indexed: 01/27/2023] Open
Abstract
The early-life microbiota triggers life-long effects on physiological functions and health disorders. Previous studies in adult twins or animal models have revealed associations between host genetics and the harmonious microbiota. However, such associations may be obscured by the fact that each intra-pair of twins will continually encounter various environmental factors as they grow up. Here, we collected the meconium samples from nineteen dizygotic pairs (DZ, n = 38) and nine monozygotic pairs (MZ, n = 18) with cesarean delivery, and 16S rRNA gene sequencing was performed to profile the microbiome at birth. Diversity analysis showed that alpha diversity was not significantly different between two groups, whereas beta diversity of MZ twins was significantly lower than that of either DZ twins or unrelated individuals (i.e., randomly selected individual pairs of non-twinship) (p < 0.05). Two groups had very similar microbial classifications but different relative abundances of certain taxa including more Firmicutes (p = 0.05, Wilcoxon test) at the phylum level and lower abundances of five genera (p < 0.05) in DZ group compared to MZ group, including Rheinheimera, Proteus, SMB53, Sphingobium, and Megamonas. Co-occurrence analysis in each group showed slightly more complicated microbial interactions in DZ than MZ twins, although 22 shared bacterial genera co-existed in two groups, with both Rheinheimera and Megamonas having different centralities in their respective co-occurrence networks. Mean intra-class correlation coefficient (ICC) were also significantly higher for MZ (0.312) compared to DZ twins (0.138) (p < 0.05). The predicted microbial gene functions related to carbohydrate were higher in DZ group, whereas folding, sorting, degradation, cell motility pathways and energy metabolism were markedly over-represented in the microbiota of MZ group. In summary, our study uncovered that microbial diversity and components of the meconium microbiome between DZ and MZ twins were partially consistent with that in singleton neonates by cesarean delivery, but several distinctions related to the heritability supported genetic contributions to intestinal microbiome in early life.
Collapse
Affiliation(s)
- Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Su Yao
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Kun Cheng
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Lili Xu
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Lingling Hou
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Huijun Feng
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xuejian Yu
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xin Zhang
- China Center of Industrial Culture Collection (CICC), China National Research Institute of Food & Fermentation Industries Co., Ltd., Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
17
|
Wong WS, Sabu P, Deopujari V, Levy S, Shah AA, Clemency N, Provenzano M, Saadoon R, Munagala A, Baker R, Baveja R, Mueller NT, Dominguez-Bello MG, Huddleston K, Niederhuber JE, Hourigan SK. Prenatal and Peripartum Exposure to Antibiotics and Cesarean Section Delivery Are Associated with Differences in Diversity and Composition of the Infant Meconium Microbiome. Microorganisms 2020; 8:E179. [PMID: 32012716 PMCID: PMC7074690 DOI: 10.3390/microorganisms8020179] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/16/2022] Open
Abstract
The meconium microbiome may provide insight into intrauterine and peripartum exposures and the very earliest intestinal pioneering microbes. Prenatal antibiotics have been associated with later obesity in children, which is thought to be driven by microbiome dependent mechanisms. However, there is little data regarding associations of prenatal or peripartum antibiotic exposure, with or without cesarean section (CS), with the features of the meconium microbiome. In this study, 16S ribosomal RNA gene sequencing was performed on bacterial DNA of meconium samples from 105 infants in a birth cohort study. After multivariable adjustment, delivery mode (p = 0.044), prenatal antibiotic use (p = 0.005) and peripartum antibiotic use (p < 0.001) were associated with beta diversity of the infant meconium microbiome. CS (vs. vaginal delivery) and peripartum antibiotics were also associated with greater alpha diversity of the meconium microbiome (Shannon and Simpson, p < 0.05). Meconium from infants born by CS (vs. vaginal delivery) had lower relative abundance of the genus Escherichia (p < 0.001). Prenatal antibiotic use and peripartum antibiotic use (both in the overall analytic sample and when restricting to vaginally delivered infants) were associated with differential abundance of several bacterial taxa in the meconium. Bacterial taxa in the meconium microbiome were also differentially associated with infant excess weight at 12 months of age, however, sample size was limited for this comparison. In conclusion, prenatal and peripartum antibiotic use along with CS delivery were associated with differences in the diversity and composition of the meconium microbiome. Whether or not these differences in the meconium microbiome portend risk for long-term health outcomes warrants further exploration.
Collapse
Affiliation(s)
- Wendy S.W. Wong
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Priya Sabu
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Varsha Deopujari
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Shira Levy
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Ankit A. Shah
- Division of Maternal Fetal Medicine, Department of Ob/Gyn, Inova Fairfax Hospital, Falls Church, VA 22042, USA;
| | - Nicole Clemency
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Marina Provenzano
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Reem Saadoon
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Akhil Munagala
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
| | - Robin Baker
- Fairfax Neonatal Associates, Falls Church, VA 22042, USA; (R.B.); (R.B.)
| | - Rajiv Baveja
- Fairfax Neonatal Associates, Falls Church, VA 22042, USA; (R.B.); (R.B.)
| | - Noel T. Mueller
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA;
| | | | - Kathi Huddleston
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
- College of Health and Human Services, George Mason University, Fairfax, VA 22030, USA
| | - John E. Niederhuber
- Inova Translational Medicine Institute, Falls Church, Inova Fairfax Hospital, Falls Church, VA 22042, USA (V.D.); (S.L.); (N.C.); (M.P.); (R.S.); (A.M.); (K.H.); (J.E.N.)
- Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Suchitra K. Hourigan
- Inova Children’s Hospital, Inova Fairfax Hospital, Falls Church, VA 22042, USA
- Pediatric Specialists of Virginia, Fairfax, VA 22031, USA
| |
Collapse
|
18
|
Celiac Disease and the Microbiome. Nutrients 2019; 11:nu11102403. [PMID: 31597349 PMCID: PMC6835875 DOI: 10.3390/nu11102403] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the hypothesis that changes in both the composition and function of the intestinal microbiome are associated with a number of chronic inflammatory diseases including celiac disease (CD). One of the major advances in the field of microbiome studies over the last few decades has been the development of culture-independent approaches to identify and quantify the components of the human microbiota. The study of nucleic acids DNA and RNA found in feces or other biological samples bypasses the need for tissue cultures and also allows the characterization of non-cultivable microbes. Current evidence on the composition of the intestinal microbiome and its role as a causative trigger for CD is highly heterogeneous and sometimes contradictory. This review is aimed at summarizing both pre-clinical (basic science data) and clinical (cross-sectional and prospective studies) evidence addressing the relationship between the intestinal microbiome and CD.
Collapse
|
19
|
Wang X, Tsai T, Deng F, Wei X, Chai J, Knapp J, Apple J, Maxwell CV, Lee JA, Li Y, Zhao J. Longitudinal investigation of the swine gut microbiome from birth to market reveals stage and growth performance associated bacteria. MICROBIOME 2019; 7:109. [PMID: 31362781 PMCID: PMC6664762 DOI: 10.1186/s40168-019-0721-7] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/05/2019] [Indexed: 05/24/2023]
Abstract
BACKGROUND Despite recent advances in the understanding of the swine gut microbiome at different growth stages, a comprehensive longitudinal study of the lifetime (birth to market) dynamics of the swine gut microbiome is lacking. RESULTS To fill in this gap of knowledge, we repeatedly collected a total of 273 rectal swabs from 18 pigs during lactation (day (d) 0, 11, 20), nursery (d 27, 33, 41, 50, 61), growing (d 76, 90, 104, 116), and finishing (d 130, 146, 159, 174) stages. DNA was extracted and subjected to sequencing with an Illumina Miseq sequencer targeting the V4 region of the 16S rRNA gene. Sequences were analyzed with the Deblur algorithm in the QIIME2 package. A total of 19 phyla were detected in the lifetime pig gut microbiome with Firmicutes and Bacteroidetes being the most abundant. Alpha diversity including community richness (e.g., number of observed features) and diversity (e.g., Shannon index) showed an overall increasing trend. Distinct shifts in microbiome structure along different growth stages were observed. LEfSe analysis revealed 91 bacterial features that are stage-specific. To validate these discoveries, we performed fecal microbiota transplantation (FMT) by inoculating weanling pigs with mature fecal microbiota from a growing stage pig. Similar stage-specific patterns in microbiome diversity and structures were also observed in both the FMT pigs and their littermates. Although FMT remarkably increased growth performance, it did not change the overall swine gut microbiome. Only a few taxa including those associated with Streptococcus and Clostridiaceae were enriched in the FMT pigs. These data, together with several other lines of evidence, indicate potential roles these taxa play in promoting animal growth performance. Diet, especially crude fiber from corn, was a major factor shaping the swine gut microbiome. The priority effect, i.e., the order and timing of species arrival, was more evident in the solid feed stages. CONCLUSIONS The distinct stage-associated swine gut microbiome may be determined by the differences in diet and/or gut physiology at different growth stages. Our study provides insight into mechanisms governing gut microbiome succession and also underscores the importance of optimizing stage-specific probiotics aimed at improving animal health and production.
Collapse
Affiliation(s)
- Xiaofan Wang
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Tsungcheng Tsai
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Feilong Deng
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan China
| | - Xiaoyuan Wei
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Jianmin Chai
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Joshua Knapp
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Jason Apple
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Charles V. Maxwell
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| | - Jung Ae Lee
- Agricultural Statistics Laboratory, University of Arkansas, Fayetteville, AR USA
| | - Ying Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan China
| | - Jiangchao Zhao
- Department of Animal Science, University of Arkansas, Fayetteville, AR USA
| |
Collapse
|
20
|
Hu J, Ly J, Zhang W, Huang Y, Glover V, Peter I, Hurd YL, Nomura Y. Microbiota of newborn meconium is associated with maternal anxiety experienced during pregnancy. Dev Psychobiol 2019; 61:640-649. [PMID: 30908632 DOI: 10.1002/dev.21837] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 12/18/2018] [Accepted: 01/13/2019] [Indexed: 12/11/2022]
Abstract
Little is known about whether a mother's psychological state during pregnancy influences her offspring's microbiome. This study examined whether maternal anxiety, depression, and stress during pregnancy is associated with the diversity of meconium microbiome, the first internal discharge, in 75 newborns from an existing birth cohort study. The meconium microbiome was profiled using multibarcode16S rRNA sequencing at V3-V4 hypervariable region followed by taxonomic assignment to the green gene 16S references at 97% similarity and diversity analysis at the genus level. Results showed that the meconium contained diversified microbiota, and greater pregnancy-related anxiety was significantly associated with a less diverse meconium microbiota community (p = 0.001). At the specific taxa level, greater pregnancy-related anxiety was correlated with a lower level of the Enterococcaceae family (p = 2e-4, Spearman rho = -0.43). These findings support a significant role of prenatal maternal mood in the early-life bacteria colonization of their offspring.
Collapse
Affiliation(s)
- Jianzhong Hu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jenny Ly
- Department of Psychology, Queens College, City University of New York, New York, New York
| | - Wei Zhang
- Department of Psychology, Queens College, City University of New York, New York, New York.,Department of Psychology, New Jersey City University, Jersey City, New Jerse, New Jersey
| | - Yonglin Huang
- Graduate Center, City University of New York, Brooklyn, New York, NY
| | - Vivette Glover
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yoko Nomura
- Department of Psychology, Queens College, City University of New York, New York, New York.,Graduate Center, City University of New York, Brooklyn, New York, NY.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
21
|
Lisowska-Myjak B, Skarżyńska E, Wilczyńska P, Jakimiuk A. Correlation between the concentrations of lactoferrin and neutrophil gelatinase-associated lipocalin in meconium. Biometals 2017; 31:123-129. [PMID: 29285663 PMCID: PMC5778183 DOI: 10.1007/s10534-017-0073-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 12/16/2017] [Indexed: 01/15/2023]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) and lactoferrin (Lf) are among the key components of the innate immune system due to their ability to bind iron with high affinity and thus control inflammation. The aim of this study was to test the use of NGAL and LF measurements in meconium for the assessment of the intrauterine homeostasis. NGAL and Lf concentrations were measured using ELISA kits in all serial meconium portions (n = 81) collected from 20 healthy neonates. Mean ± SD meconium concentration of Lf was 45.07 ± 78.53 µg/g and more than 1000-fold higher compared with that of NGAL at 1.93 ± 2.46 ng/g. The correlation between the two proteins (r = 0.83, p < 0.0001) was found only for portions with Lf concentrations > 25 μg/g. High variability of NGAL and Lf concentrations in meconium and their correlations prove their key role as biomarkers of the fetal condition in utero. NGAL and Lf measured in meconium are candidate biomarkers for fetal iron status.
Collapse
Affiliation(s)
- Barbara Lisowska-Myjak
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, ul. Banacha 1, 02-097, Warsaw, Poland.
| | - Ewa Skarżyńska
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, ul. Banacha 1, 02-097, Warsaw, Poland
| | - Paulina Wilczyńska
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, ul. Banacha 1, 02-097, Warsaw, Poland
| | - Artur Jakimiuk
- Clinical Department of Obstetrics, Female Diseases and Gynecological Oncology, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| |
Collapse
|