101
|
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
|
102
|
Chen JF, Ou-Yang MC, Hsia KC, Li CM, Yeh YT, Ho HH. A Three-Arm, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 Used Individually in Healthy Infants. Nutrients 2023; 15:3426. [PMID: 37571365 PMCID: PMC10421338 DOI: 10.3390/nu15153426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Probiotics are considered safe and beneficial to human health. However, the safety of Lactobacillus salivarius AP-32 and Bifidobacterium animalis CP-9 in infants has not been confirmed. This study was to assess the safety of long-term oral administration of L. salivarius AP-32 and B. animalis CP-9 in healthy infants compared with placebo. A three-arm, randomized, double-blind, placebo-controlled trial was conducted in healthy, full-term infants. Eighty-eight infants between 7 days and 2 months (60 ± 7 days) of age were selected and randomized to treatment with L. salivarius AP-32, B. animalis CP-9 or placebo for 4 months. The unblinding indicated subjects were randomized to receive B. animalis CP-9 (N = 28), L. salivarius AP-32 (N = 29), or placebo (N = 31). A total of 76 infants completed the 4-month treatment with fully compliance. The primary outcome was weight gain, with no significant difference in infant weight at 4 months when comparing AP-32 or CP-9 group with the placebo group, either. The head circumference and recumbent length of the CP-9 group were not significantly different from those of the placebo group. The recumbent length of the AP-32 group was slightly lower than that in the placebo group at month 4, but there was no difference between the two groups in head circumference. Overall, the growth trend of all treatments was similar without significant difference. Furthermore, there were no apparent differences between each group in digestive tolerance, the occurrence of adverse events, crying/fussing time and episodes, alpha diversity, and beta diversity. The CP-9 group showed a significant increase in the abundance of the Bacteroides genus, while the AP-32 group demonstrated a significant increase in the abundance of the Lactobacillus genus when comparing the two probiotic groups. Our study findings indicate that the oral administration of both AP-32 and CP-9 strains has a positive impact on the maintenance of a healthy gut flora in infants. Long-term use of L. salivarius AP-32 or B. animalis CP-9 is safe for infants from 7 days to 6 months of age.
Collapse
Affiliation(s)
- Jui-Fen Chen
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Mei-Chen Ou-Yang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Ko-Chiang Hsia
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | | | - Ching-Min Li
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung 831, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Fooyin University, Kaohsiung 831, Taiwan
| | - Hsieh-Hsun Ho
- Department of Research and Design, Glac Biotech Co., Ltd., Tainan 744, Taiwan; (J.-F.C.); (K.-C.H.); (C.-M.L.)
| |
Collapse
|
103
|
Blackmer-Raynolds L, Sampson TR. Overview of the Gut Microbiome. Semin Neurol 2023; 43:518-529. [PMID: 37562449 DOI: 10.1055/s-0043-1771463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
The human gastrointestinal tract is home to trillions of microorganisms-collectively referred to as the gut microbiome-that maintain a symbiotic relationship with their host. This diverse community of microbes grows and changes as we do, with developmental, lifestyle, and environmental factors all shaping microbiome community structure. Increasing evidence suggests this relationship is bidirectional, with the microbiome also influencing host physiological processes. For example, changes in the gut microbiome have been shown to alter neurodevelopment and have lifelong effects on the brain and behavior. Age-related changes in gut microbiome composition have also been linked to inflammatory changes in the brain, perhaps increasing susceptibility to neurological disease. Indeed, associations between gut dysbiosis and many age-related neurological diseases-including Parkinson's disease, Alzheimer's disease, multiple sclerosis, and amyotrophic lateral sclerosis-have been reported. Further, microbiome manipulation in animal models of disease highlights a potential role for the gut microbiome in disease development and progression. Although much remains unknown, these associations open up an exciting new world of therapeutic targets, potentially allowing for improved quality of life for a wide range of patient populations.
Collapse
Affiliation(s)
| | - Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
104
|
Panzer AR, Sitarik AR, Fadrosh D, Havstad SL, Jones K, Davidson B, Finazzo S, Wegienka GR, Woodcroft K, Lukacs NW, Levin AM, Ownby DR, Johnson CC, Lynch SV, Zoratti EM. The impact of prenatal dog keeping on infant gut microbiota development. Clin Exp Allergy 2023; 53:833-845. [PMID: 36916778 PMCID: PMC11163251 DOI: 10.1111/cea.14303] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/15/2023]
Abstract
INTRODUCTION Prenatal and early-life dog exposure has been linked to reduced childhood allergy and asthma. A potential mechanism includes altered early immune development in response to changes in the gut microbiome among dog-exposed infants. We thus sought to determine whether infants born into homes with indoor dog(s) exhibit altered gut microbiome development. METHODS Pregnant women living in homes with dogs or in pet-free homes were recruited in southeast Michigan. Infant stool samples were collected at intervals between 1 week and 18 months after birth and microbiome was assessed using 16S ribosomal sequencing. Perinatal maternal vaginal/rectal swabs and stool samples were sequenced from a limited number of mothers. Mixed effect adjusted models were used to assess stool microbial community trajectories comparing infants from dog-keeping versus pet-free homes with adjustment for relevant covariates. RESULTS Infant gut microbial composition among vaginally born babies became less similar to the maternal vaginal/rectal microbiota and more similar to the maternal gut microbiota with age-related accumulation of bacterial species with advancing age. Stool samples from dog-exposed infants were microbially more diverse (p = .041) through age 18 months with enhanced diversity most apparent between 3 and 6 months of age. Statistically significant effects of dog exposure on β-diversity metrics were restricted to formula-fed children. Across the sample collection period, dog exposure was associated with Fusobacterium genera enrichment, as well as enrichment of Collinsella, Ruminococcus, Clostridaceae and Lachnospiraceae OTUs. CONCLUSION Prenatal/early-life dog exposure is associated with an altered gut microbiome during infancy and supports a potential mechanism explaining lessened atopy and asthma risk. Further research directly linking specific dog-attributable changes in the infant gut microbiome to the risk of allergic disorders is needed.
Collapse
Affiliation(s)
- Ariane R Panzer
- Department of Medicine, University of California, San Francisco, California, USA
| | - Alexandra R Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Doug Fadrosh
- Department of Medicine, University of California, San Francisco, California, USA
| | - Suzanne L Havstad
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Kyra Jones
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Brent Davidson
- Department of Women's Health, Henry Ford Health System, Detroit, Michigan, USA
| | - Salvatore Finazzo
- Department of Obstetrics and Gynecology, Henry Ford Wyandotte Hospital, Wyandotte, Michigan, USA
| | - Ganesa R Wegienka
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Kimberley Woodcroft
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
- Center for Bioinformatics, Henry Ford Health System, Detroit, Michigan, USA
| | - Dennis R Ownby
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
- Department of Pediatrics, Augusta University, Augusta, Georgia, USA
| | - Christine C Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Susan V Lynch
- Department of Medicine, University of California, San Francisco, California, USA
| | - Edward M Zoratti
- Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
105
|
Tarracchini C, Alessandri G, Fontana F, Rizzo SM, Lugli GA, Bianchi MG, Mancabelli L, Longhi G, Argentini C, Vergna LM, Anzalone R, Viappiani A, Turroni F, Taurino G, Chiu M, Arboleya S, Gueimonde M, Bussolati O, van Sinderen D, Milani C, Ventura M. Genetic strategies for sex-biased persistence of gut microbes across human life. Nat Commun 2023; 14:4220. [PMID: 37452041 PMCID: PMC10349097 DOI: 10.1038/s41467-023-39931-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
Although compositional variation in the gut microbiome during human development has been extensively investigated, strain-resolved dynamic changes remain to be fully uncovered. In the current study, shotgun metagenomic sequencing data of 12,415 fecal microbiomes from healthy individuals are employed for strain-level tracking of gut microbiota members to elucidate its evolving biodiversity across the human life span. This detailed longitudinal meta-analysis reveals host sex-related persistence of strains belonging to common, maternally-inherited species, such as Bifidobacterium bifidum and Bifidobacterium longum subsp. longum. Comparative genome analyses, coupled with experiments including intimate interaction between microbes and human intestinal cells, show that specific bacterial glycosyl hydrolases related to host-glycan metabolism may contribute to more efficient colonization in females compared to males. These findings point to an intriguing ancient sex-specific host-microbe coevolution driving the selective persistence in women of key microbial taxa that may be vertically passed on to the next generation.
Collapse
Affiliation(s)
- Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Alessandri
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Federico Fontana
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- GenProbio srl, Parma, Italy
| | - Sonia Mirjam Rizzo
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Massimiliano Giovanni Bianchi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Laura Maria Vergna
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | | | | | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Martina Chiu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, CSIC, 33300, Villaviciosa, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, CSIC, 33300, Villaviciosa, Spain
| | - Ovidio Bussolati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, T12YT20, Cork, Ireland
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy.
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy.
| |
Collapse
|
106
|
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: 1.0] [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
- 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
| | - Smaragdi Fessatou
- Third Department of Pediatrics, "Attikon" General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece
| | - 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
- 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
|
107
|
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: 2.0] [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
|
108
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
109
|
Tejesvi MV, Turunen J, Salmi S, Reunanen J, Paalanne N, Tapiainen T. Delivery Mode and Perinatal Antibiotics Influence the Infant Gut Bacteriome and Mycobiome: A Network Analysis. J Fungi (Basel) 2023; 9:718. [PMID: 37504707 PMCID: PMC10381809 DOI: 10.3390/jof9070718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Both exposure to antibiotics at birth and delivery via Caesarean section influence the gut bacteriome's development in infants. Using 16S rRNA and internal transcribed spacer sequencing on the Ion Torrent platform, we employed network analysis to investigate the bacterial and fungal interkingdom relationships in the gut microbiome from birth to age 18 months in a prospective cohort study of 140 infants. The gut microbiome at ages six and 18 months revealed distinctive microbial interactions, including both positive and negative associations between bacterial and fungal genera in the gut ecosystem. Perinatal factors, delivery mode and intrapartum antibiotic exposure affected the associations between bacterial and fungal species. In infants exposed and unexposed to perinatal antibiotics, the gut microbiome formed distinct networks for the bacteriome and mycobiome. The fungi Saccharomyces, Trichosporon, Pezoloma, Cystofilobasidium, Rigidoporus and Fomitopsis were strongly associated with exposure to antibiotics at birth. Hyaloscypha, Trichosporon, Fomitopsis and Vishniacozyma were strongly associated with the control group that was not exposed to antibiotics. Five distinct networks were formed according to delivery mode. The present study confirms that bacteria and fungi clearly interact in the infant gut ecosystem. Furthermore, perinatal factors appear to influence the relationships between bacteria and fungi in the developing gut microbiome.
Collapse
Affiliation(s)
- Mysore V Tejesvi
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
- Ecology and Genetics, Faculty of Science, University of Oulu, 90014 Oulu, Finland
| | - Jenni Turunen
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
- Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
| | - Sonja Salmi
- Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, 90014 Oulu, Finland
- Disease Networks Research Unit, University of Oulu, 90014 Oulu, Finland
| | - Justus Reunanen
- Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
- Research Unit of Translational Medicine, University of Oulu, 90014 Oulu, Finland
| | - Niko Paalanne
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, 90014 Oulu, Finland
| | - Terhi Tapiainen
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, 90014 Oulu, Finland
| |
Collapse
|
110
|
Fan D, Xia Q, Lin D, Ma Y, Rao J, Liu L, Tang H, Xu T, Li P, Chen G, Zhou Z, Guo X, Zhang Z, Liu Z. Role of breastfeeding on maternal and childhood cancers: An umbrella review of meta-analyses. J Glob Health 2023; 13:04067. [PMID: 37350093 PMCID: PMC10288171 DOI: 10.7189/jogh.13.04067] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023] Open
Abstract
Background Multiple studies and meta-analyses have claimed that breastfeeding is inversely correlated with maternal and childhood cancers. These results could either be causal or confounded by shared risk factors. By conducting an umbrella review, we aimed to consolidate the relationship between breastfeeding and maternal and childhood cancers. Methods We searched PubMed, Embase, Web of Science, Elsevier ScienceDirect, and Cochrane Library databases from inception to December 2022. Two reviewers independently extracted the data and assessed the quality of the studies using standardised forms. We considered two types of breastfeeding comparisons ("ever" vs "never" breastfeeding; and "longest" vs "shortest" duration). We estimated the pooled risk and 95% confidence interval (CI) for each meta-analysis. Results We included seventeen meta-analyses with 55 comparisons. There was an inverse correlation between breastfeeding and childhood leukaemia (pooled risk = 0.90, 95% CI = 0.81-0.99), neuroblastoma (pooled risk = 0.81, 95% CI = 0.71-0.93), maternal ovarian cancer (pooled risk = 0.76, CI = 0.71-0.81), breast cancer (pooled risk = 0.85, 95% CI = 0.82-0.88), and oesophageal cancer (pooled risk = 0.67, 95% CI = 0.54-0.81) for "ever" vs "never" breastfeeding; and with childhood leukaemia (pooled risk = 0.94, 95% CI = 0.89-0.98), and maternal ovarian cancer (pooled risk = 0.84, 95% CI = 0.78-0.90) and breast cancer (pooled risk = 0.92, 95% CI = 0.89-0.96) for "longest" vs "shortest" breastfeeding duration. Conclusions We found evidence that breastfeeding may reduce the risk of maternal breast cancer, ovarian cancers, and childhood leukaemia, suggesting positive implications for influencing women's decision in breastfeeding. Registration PROSPERO (CRD42021255608).
Collapse
Affiliation(s)
- Dazhi Fan
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
- Department of Obstetrics, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Qing Xia
- Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, School of Public Health & Social Work, Faculty of Health, Queensland University of Technology, Australia
| | - Dongxin Lin
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Yubo Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jiaming Rao
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Li Liu
- Department of Library, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hai Tang
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Tingting Xu
- Department of Health Management and Policy, School of Public Health, Capital Medical University, Beijing, China
| | - Pengsheng Li
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Gengdong Chen
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Zixing Zhou
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Xiaoling Guo
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
- Department of Obstetrics, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Zhifang Zhang
- Department of Obstetrics, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Zhengping Liu
- Foshan Fetal Medicine Research Institute, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
- Department of Obstetrics, Affiliated Foshan Women and Children Hospital, Southern Medical University, Foshan, Guangdong, China
| |
Collapse
|
111
|
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: 15.0] [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
|
112
|
Graspeuntner S, Lupatsii M, Dashdorj L, Rody A, Rupp J, Bossung V, Härtel C. First-Day-of-Life Rectal Swabs Fail To Represent Meconial Microbiota Composition and Underestimate the Presence of Antibiotic Resistance Genes. Microbiol Spectr 2023; 11:e0525422. [PMID: 37097170 PMCID: PMC10269712 DOI: 10.1128/spectrum.05254-22] [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: 12/22/2022] [Accepted: 04/10/2023] [Indexed: 04/26/2023] Open
Abstract
The human gut microbiome plays a vital role in health and disease. In particular, the first days of life provide a unique window of opportunity for development and establishment of microbial community. Currently, stool samples are known to be the most widely used sampling approach for studying the gut microbiome. However, complicated sample acquisition at certain time points, challenges in transportation, and patient discomfort underline the need for development of alternative sampling approaches. One of the alternatives is rectal swabs, shown to be a reliable proxy for gut microbiome analysis when obtained from adults. Here, we compare the usability of rectal swabs and meconium paired samples collected from infants on the first days of life. Our results indicate that the two sampling approaches display significantly distinct patterns in microbial composition and alpha and beta diversity as well as detection of resistance genes. Moreover, the dissimilarity between the two collection methods was greater than the interindividual variation. Therefore, we conclude that rectal swabs are not a reliable proxy compared to stool samples for gut microbiome analysis when collected on the first days of a newborn's life. IMPORTANCE Currently, there are numerous suggestions on how to ease the notoriously complex and error-prone methodological setups to study the gut microbiota of newborns during the first days of life. Especially, meconium samples are regularly failing to yield meaningful data output and therefore have been suggested to be replaced by rectal swabs as done in adults as well. We find this development toward a simplified method to be producing dramatically erroneous results, skewing data interpretation away from the real aspects to be considered for neonatal health during the first days of life. We have put together our knowledge on this critical aspect with careful consideration and identified the failure of rectal swabs to be a replacement for sampling of meconium in term-born newborns.
Collapse
Affiliation(s)
- S. Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - M. Lupatsii
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - L. Dashdorj
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - A. Rody
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - J. Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - V. Bossung
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - C. Härtel
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
113
|
Artzi-Medvedik R, Mariani I, Valente EP, Lazzerini M, Chertok IA. Factors associated with exclusive breastfeeding in Israel during the COVID-19 pandemic: a subset of the IMAgiNE EURO cross-sectional study. Int Breastfeed J 2023; 18:30. [PMID: 37296409 PMCID: PMC10250857 DOI: 10.1186/s13006-023-00568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Evidence has shown that restrictions during the COVID-19 pandemic have negatively affected breastfeeding support and outcomes in hospitals in many countries. The aims of the study were to describe exclusive breastfeeding rates and identify factors associated with exclusive breastfeeding at hospital discharge among women who gave birth during the COVID-19 pandemic in Israel. METHODS A cross-sectional online anonymous survey based on WHO standards for improving quality of maternal and newborn care in health facilities was conducted among a sample of women who gave birth to a healthy singleton infant in Israel during the pandemic (between March 2020 and April 2022). The socio-ecological approach was employed to examine intrapersonal, interpersonal, organizational, and community/society factors associated with exclusive breastfeeding at hospital discharge according to women perspectives. RESULTS Among the 235 Israeli participants, 68.1% exclusively breastfed, 27.7% partially breastfed, and 4.2% did not breastfeed at discharge. Results of the adjusted logistic regression model showed that factors significantly associated with exclusive breastfeeding were the intrapersonal factor of multiparity (adjusted OR 2.09; 95% Confidence Interval 1.01,4.35) and the organizational factors of early breastfeeding in the first hour (aOR 2.17; 95% CI 1.06,4.45), and rooming-in (aOR 2.68; 95% CI 1.41,5.07). CONCLUSIONS Facilitating early breastfeeding initiation and supporting rooming-in are critical to promoting exclusive breastfeeding. These factors, reflecting hospital policies and practices, along with parity, are significantly associated with breastfeeding outcomes and highlight the influential role of the maternity environment during the COVID-19 pandemic. Maternity care in hospitals should follow evidence-based breastfeeding recommendations also during the pandemic, promoting early exclusive breastfeeding and rooming-in among all women, with particular attention to providing lactation support to primiparous women. TRIAL REGISTRATION Clinical Trials NCT04847336.
Collapse
Affiliation(s)
- Rada Artzi-Medvedik
- School of Nursing, College of Health Sciences and Professions, Ohio University, Athens, OH, USA
| | - Ilaria Mariani
- WHO Collaborating Centre for Maternal and Child Health, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Emanuelle Pessa Valente
- WHO Collaborating Centre for Maternal and Child Health, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy.
| | - Marzia Lazzerini
- WHO Collaborating Centre for Maternal and Child Health, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
- Maternal Adolescent Reproductive and Child Health Care Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Ilana Azulay Chertok
- School of Nursing, College of Health Sciences and Professions, Ohio University, Athens, OH, USA
| |
Collapse
|
114
|
Pace E, Yanowitz TD, Waltz P, Morowitz MJ. Antibiotic therapy and necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151308. [PMID: 37295297 DOI: 10.1016/j.sempedsurg.2023.151308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
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
|
115
|
Wu Y, Zhang G, Wang Y, Wei X, Liu H, Zhang L, Zhang L. A Review on Maternal and Infant Microbiota and Their Implications for the Prevention and Treatment of Allergic Diseases. Nutrients 2023; 15:nu15112483. [PMID: 37299446 DOI: 10.3390/nu15112483] [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: 04/26/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Allergic diseases, which are closely related to the composition and metabolism of maternal and infant flora, are prevalent in infants worldwide. The mother's breast milk, intestinal, and vaginal flora directly or indirectly influence the development of the infant's immune system from pregnancy to lactation, and the compositional and functional alterations of maternal flora are associated with allergic diseases in infants. Meanwhile, the infant's own flora, represented by the intestinal flora, indicates and regulates the occurrence of allergic diseases and is altered with the intervention of allergic diseases. By searching and selecting relevant literature in PubMed from 2010 to 2023, the mechanisms of allergy development in infants and the links between maternal and infant flora and infant allergic diseases are reviewed, including the effects of flora composition and its consequences on infant metabolism. The critical role of maternal and infant flora in allergic diseases has provided a window for probiotics as a microbial therapy. Therefore, the uses and mechanisms by which probiotics, such as lactic acid bacteria, can help to improve the homeostasis of both the mother and the infant, and thereby treat allergies, are also described.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yucong Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin Wei
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huanhuan Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
116
|
Kouete MT, Bletz MC, LaBumbard BC, Woodhams DC, Blackburn DC. Parental care contributes to vertical transmission of microbes in a skin-feeding and direct-developing caecilian. Anim Microbiome 2023; 5:28. [PMID: 37189209 PMCID: PMC10184399 DOI: 10.1186/s42523-023-00243-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/20/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Our current understanding of vertebrate skin and gut microbiomes, and their vertical transmission, remains incomplete as major lineages and varied forms of parental care remain unexplored. The diverse and elaborate forms of parental care exhibited by amphibians constitute an ideal system to study microbe transmission, yet investigations of vertical transmission among frogs and salamanders have been inconclusive. In this study, we assess bacteria transmission in Herpele squalostoma, an oviparous direct-developing caecilian in which females obligately attend juveniles that feed on their mother's skin (dermatophagy). RESULTS We used 16S rRNA amplicon-sequencing of the skin and gut of wild caught H. squalostoma individuals (males, females, including those attending juveniles) as well as environmental samples. Sourcetracker analyses revealed that juveniles obtain an important portion of their skin and gut bacteria communities from their mother. The contribution of a mother's skin to the skin and gut of her respective juveniles was much larger than that of any other bacteria source. In contrast to males and females not attending juveniles, only the skins of juveniles and their mothers were colonized by bacteria taxa Verrucomicrobiaceae, Nocardioidaceae, and Erysipelotrichaceae. In addition to providing indirect evidence for microbiome transmission linked to parental care among amphibians, our study also points to noticeable differences between the skin and gut communities of H. squalostoma and that of many frogs and salamanders, which warrants further investigation. CONCLUSION Our study is the first to find strong support for vertical bacteria transmission attributed to parental care in a direct-developing amphibian species. This suggests that obligate parental care may promote microbiome transmission in caecilians.
Collapse
Affiliation(s)
- Marcel T Kouete
- School of Natural Resources and Environment, University of Florida, Gainesville, FL, 32611, USA.
- Department of Natural History, Florida Museum of Natural History, University of Florida, Gainesville, FL, 32611, USA.
| | - Molly C Bletz
- Department of Biology, University of Massachusetts, Boston, MA, 02125, USA
| | | | - Douglas C Woodhams
- Department of Biology, University of Massachusetts, Boston, MA, 02125, USA
| | - David C Blackburn
- Department of Natural History, Florida Museum of Natural History, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
117
|
Baloh CH, Venturi GM, Fischer BM, Sadder LS, Kim-Chang JJ, Chan C, De Paris K, Yin L, Aldrovandi GM, Goodenow MM, Sleasman JW. Biomarkers detected in cord blood predict vaccine responses in young infants. Front Immunol 2023; 14:1152538. [PMID: 37251388 PMCID: PMC10213698 DOI: 10.3389/fimmu.2023.1152538] [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: 01/27/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Factors influencing vaccine immune priming in the first year of life involve both innate and adaptive immunity but there are gaps in understanding how these factors sustain vaccine antibody levels in healthy infants. The hypothesis was that bioprofiles associated with B cell survival best predict sustained vaccine IgG levels at one year. Methods Longitudinal study of plasma bioprofiles in 82 term, healthy infants, who received standard recommended immunizations in the United States, with changes in 15 plasma biomarker concentrations and B cell subsets associated with germinal center development monitored at birth, soon after completion of the initial vaccine series at 6 months, and prior to the 12-month vaccinations. Post vaccination antibody IgG levels to Bordetella pertussis, tetanus toxoid, and conjugated Haemophilus influenzae type B (HiB) were outcome measures. Results Using a least absolute shrinkage and selection operator (lasso) regression model, cord blood (CB) plasma IL-2, IL-17A, IL-31, and soluble CD14 (sCD14) were positively associated with pertussis IgG levels at 12 months, while CB plasma concentrations of APRIL and IL-33 were negatively associated. In contrast, CB concentrations of sCD14 and APRIL were positively associated with sustained tetanus IgG levels. A separate cross-sectional analysis of 18 mother/newborn pairs indicated that CB biomarkers were not due to transplacental transfer, but rather due to immune activation at the fetal/maternal interface. Elevated percentages of cord blood switched memory B cells were positively associated with 12-month HiB IgG levels. BAFF concentrations at 6 and 12 months were positively associated with pertussis and HiB IgG levels respectively. Discussion Sustained B cell immunity is highly influenced by early life immune dynamics beginning prior to birth. The findings provide important insights into how germinal center development shapes vaccine responses in healthy infants and provide a foundation for studies of conditions that impair infant immune development.
Collapse
Affiliation(s)
- Carolyn H. Baloh
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Guglielmo M. Venturi
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Bernard M. Fischer
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Liane S. Sadder
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Julie J. Kim-Chang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Kristina De Paris
- Institute of Global Health and Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Li Yin
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, University of California, Los Angeles, CA, United States
| | - Maureen M. Goodenow
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - John W. Sleasman
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
118
|
Kamau E, Yang S. Metagenomic Sequencing of Positive Blood Culture Fluid for Accurate Bacterial and Fungal Species Identification: A Pilot Study. Microorganisms 2023; 11:1259. [PMID: 37317232 DOI: 10.3390/microorganisms11051259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
With blood stream infections (BSIs) representing a major cause of mortality and morbidity worldwide, blood cultures play a crucial role in diagnosis, but their clinical application is dampened by the long turn-around time and the detection of only culturable pathogens. In this study, we developed and validated a shotgun metagenomics next-generation sequencing (mNGS) test directly from positive blood culture fluid, allowing for the identification of fastidious or slow growing microorganisms more rapidly. The test was built based on previously validated next-generation sequencing tests, which rely on several key marker genes for bacterial and fungal identification. The new test utilizes an open-source metagenomics CZ-ID platform for the initial analysis to generate the most likely candidate species, which is then used as a reference genome for downstream, confirmatory analysis. This approach is innovative because it takes advantage of an open-source software's agnostic taxonomic calling capability while still relying on the more established and previously validated marker gene-based identification scheme, increasing the confidence in the final results. The test showed high accuracy (100%, 30/30) for both bacterial and fungal microorganisms. We further demonstrated its clinical utility especially for anaerobes and mycobacteria that are either fastidious, slow growing, or unusual. Although applicable in only limited settings, the Positive Blood Culture mNGS test provides an incremental improvement in solving the unmet clinical needs for the diagnosis of challenging BSIs.
Collapse
Affiliation(s)
- Edwin Kamau
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Shangxin Yang
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
119
|
Christopoulou I, Kostopoulou E, Matzarapi K, Chasapi SA, Spyroulias GA, Varvarigou A. Identification of Novel Biomarkers in Late Preterm Neonates with Respiratory Distress Syndrome (RDS) Using Urinary Metabolomic Analysis. Metabolites 2023; 13:metabo13050644. [PMID: 37233686 DOI: 10.3390/metabo13050644] [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: 04/11/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Urine metabolomics is gaining traction as a means of identifying metabolic signatures associated with health and disease states. Thirty-one (31) late preterm (LP) neonates admitted to the neonatal intensive care unit (NICU) and 23 age-matched healthy LPs admitted to the maternity ward of a tertiary hospital were included in the study. Proton nuclear magnetic resonance (1H NMR) spectroscopy was employed for urine metabolomic analysis on the 1st and 3rd days of life of the neonates. The data were analyzed using univariate and multivariate statistical analysis. A unique metabolic pattern of enhanced metabolites was identified in the NICU-admitted LPs from the 1st day of life. Metabolic profiles were distinct in LPs presenting with respiratory distress syndrome (RDS). The discrepancies likely reflect differences in the gut microbiota, either due to variations in nutrient intake or as a result of medical interventions, such as the administration of antibiotics and other medications. Altered metabolites could potentially serve as biomarkers for identifying critically ill LP neonates or those at high risk for adverse outcomes later in life, including metabolic risks. The discovery of novel biomarkers may uncover potential targets for drug discovery and optimal periods for effective intervention, offering a personalized approach.
Collapse
Affiliation(s)
- Irene Christopoulou
- Department of Paediatrics, University of Patras Medical School, General University Hospital, 26500 Patras, Greece
| | - Eirini Kostopoulou
- Department of Paediatrics, University of Patras Medical School, General University Hospital, 26500 Patras, Greece
| | | | | | | | - Anastasia Varvarigou
- Department of Paediatrics, University of Patras Medical School, General University Hospital, 26500 Patras, Greece
| |
Collapse
|
120
|
Gray AN, DeFilipp Z. Fecal Microbiota Transplantation for Acute Graft-versus-Host Disease After Allogeneic Hematopoietic Cell Transplantation: Expanding the Horizon into Pediatrics. Transplant Cell Ther 2023:S2666-6367(23)01289-7. [PMID: 37169290 DOI: 10.1016/j.jtct.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
The microbiome plays a vital role in maintaining homeostasis of the intestinal microenvironment and immune response in allogeneic hematopoietic cell transplantation (HCT) recipients. Disruption of the intestinal microbiome has been associated with the development of acute graft-versus-host disease (GVHD) of the lower GI tract and worse survival. Fecal microbiota transplantation (FMT) can achieve clinical responses in refractory GVHD, establishing the promise of microbiome-directed interventions in this population. While most data about microbial changes in HCT recipients have been generated from the adult population, children with refractory GVHD represent an important group that may benefit from FMT. In this review, we first highlight characteristics that distinguish the pediatric intestinal microbiome from adults. Subsequently, we explore multiple clinical factors that warrant careful consideration to optimize the application of FMT and other microbiome-directed therapeutics to children.
Collapse
Affiliation(s)
- Ashley N Gray
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, Los Angeles, CA, USA.
| | - Zachariah DeFilipp
- Hematopoieitic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
121
|
Melekoglu E, Yılmaz B, Çevik A, Gökyıldız Sürücü Ş, Avcıbay Vurgeç B, Gözüyeşil E, Sharma H, Boyan N, Ozogul F. The Impact of the Human Milk Microbiota in the Prevention of Disease and Infant Health. Breastfeed Med 2023. [PMID: 37140562 DOI: 10.1089/bfm.2022.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Background: Human milk is recognized as an ideal food for newborns and infants owing to the presence of various nutritive factors, including healthy bacteria. Aim/Objective: This review aimed to understand the effects of human milk microbiota in both the prevention of disease and the health of infants. Methods: Data were obtained from PubMed, Scopus, Web of Science, clinical trial registries, Dergipark, and Türk Atıf Dizini up to February 2023 without language restrictions. Results: It is considered that the first human milk microbiota ingested by the newborn creates the initial microbiome of the gut system, which in turn influences the development and maturation of immunity. Bacteria present in human milk modulate the anti-inflammatory response by releasing certain cytokines, protecting the newborn against certain infections. Therefore, certain bacterial strains isolated from human milk could serve as potential probiotics for various therapeutic applications. Conclusions: In this review, the origin and significance of human milk bacteria have been highlighted along with certain factors influencing the composition of human milk microbiota. In addition, it also summarizes the health benefits of human milk as a protective agent against certain diseases and ailments.
Collapse
Affiliation(s)
- Ebru Melekoglu
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Birsen Yılmaz
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | - Ayseren Çevik
- Department of Midwifery, Cukurova University, Adana, Turkey
| | | | | | - Ebru Gözüyeşil
- Department of Midwifery, Cukurova University, Adana, Turkey
| | - Heena Sharma
- Food Technology Lab, Dairy Technology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Neslihan Boyan
- Department of Anatomy, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana, Turkey
| |
Collapse
|
122
|
Huang C, Tan H, Song M, Liu K, Liu H, Wang J, Shi Y, Hou F, Zhou Q, Huang R, Shen B, Lin X, Qin X, Zhi F. Maternal Western diet mediates susceptibility of offspring to Crohn's-like colitis by deoxycholate generation. MICROBIOME 2023; 11:96. [PMID: 37131223 PMCID: PMC10155335 DOI: 10.1186/s40168-023-01546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 04/07/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND The Western dietary pattern, characterized by high consumption of fats and sugars, has been strongly associated with an increased risk of developing Crohn's disease (CD). However, the potential impact of maternal obesity or prenatal exposure to a Western diet on offspring's susceptibility to CD remains unclear. Herein, we investigated the effects and underlying mechanisms of a maternal high-fat/high-sugar Western-style diet (WD) on offspring's susceptibility to 2,4,6-Trinitrobenzenesulfonic acid (TNBS)-induced Crohn's-like colitis. METHODS Maternal dams were fed either a WD or a normal control diet (ND) for eight weeks prior to mating and continued throughout gestation and lactation. Post-weaning, the offspring were subjected to WD and ND to create four groups: ND-born offspring fed a normal diet (N-N) or Western diet (N-W), and WD-born offspring fed a normal (W-N) or Western diet (W-W). At eight weeks of age, they were administered TNBS to induce a CD model. RESULTS Our findings revealed that the W-N group exhibited more severe intestinal inflammation than the N-N group, as demonstrated by a lower survival rate, increased weight loss, and a shorter colon length. The W-N group displayed a significant increase in Bacteroidetes, which was accompanied by an accumulation of deoxycholic acid (DCA). Further experimentation confirmed an increased generation of DCA in mice colonized with gut microbes from the W-N group. Moreover, DCA administration aggravated TNBS-induced colitis by promoting Gasdermin D (GSDMD)-mediated pyroptosis and IL-1beta (IL-1β) production in macrophages. Importantly, the deletion of GSDMD effectively restrains the effect of DCA on TNBS-induced colitis. CONCLUSIONS Our study demonstrates that a maternal Western-style diet can alter gut microbiota composition and bile acid metabolism in mouse offspring, leading to an increased susceptibility to CD-like colitis. These findings highlight the importance of understanding the long-term consequences of maternal diet on offspring health and may have implications for the prevention and management of Crohn's disease. Video Abstract.
Collapse
Affiliation(s)
- Chongyang Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huishi Tan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengyao Song
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbin Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanqiang Shi
- Institute of Dermatology and Venereology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Fengyi Hou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruo Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binghai Shen
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinlong Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoming Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
123
|
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: 42] [Impact Index Per Article: 42.0] [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
|
124
|
Wang K, Xia X, Sun L, Wang H, Li Q, Yang Z, Ren J. Microbial Diversity and Correlation between Breast Milk and the Infant Gut. Foods 2023; 12:foods12091740. [PMID: 37174279 PMCID: PMC10178105 DOI: 10.3390/foods12091740] [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: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The gut microbiota is significant for infants to grow and develop in the early stages of life. The breast milk microbiota directly or indirectly influences colonizing and the development of early infant intestinal microbiota. Therefore, we wanted to study the microbial diversity and correlation between breast milk and the infant gut. By sequencing the 16S rRNA V3-V4 regions of microbiome in infant feces 1, 14, 20, 30, and 90 days after delivery as well as those in breast milk using Illumina NovaSeq, we studied the component of microbiome in both human milk and infant stools, analyzed the diversity of microbiota, and explored the relationship between them. We found that the richest bacteria in breast milk were Acinetobacter, Stenotrophomonas, Sphingopyxis, Pseudomonas, and Streptococcus, with a small amount of Lactobacillus, Bifidobacterium, and Klebsiella. The infant feces were abundant in Bifidobacterium, Escherichia-Shigella, Klebsiella, Streptococcus, Serratia, Bacteroides, and Lactobacillus, with a small number of Acinetobacter and Pseudomonas. Acinetobacter, Bifidobacterium, Klebsiella, and Lactobacillus appeared in the breast milk and infant feces, suggesting that they were transferred from the breast milk to the infant's gut.
Collapse
Affiliation(s)
- Kaili Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiufang Xia
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lina Sun
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Hui Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Qiu Li
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhuo Yang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jing Ren
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
125
|
Oddi S, Mantziari A, Huber P, Binetti A, Salminen S, Collado MC, Vinderola G. Human Milk Microbiota Profile Affected by Prematurity in Argentinian Lactating Women. Microorganisms 2023; 11:microorganisms11041090. [PMID: 37110513 PMCID: PMC10145235 DOI: 10.3390/microorganisms11041090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
To study (16S rRNA-sequencing) the impact of gestational and corrected ages on the microbiota profile of human milk (HM) of mothers that delivered full-term and pre-term children, HM samples were obtained and classified according to the gestational age as group T (full-term births ≥37 weeks), and group P (pre-term births <37 weeks). Group P was longitudinally followed, and the samples were collected at the full-term corrected gestational age: when the chronological age plus the gestational age were ≥37 weeks (PT group). The HM microbiota composition differed depending on the gestational age (T vs. P). Group T had lower levels of Staphylococcus and higher levels of Rothia and Streptococcus, as compared to group P. The alpha Simpson diversity value was higher in group T than in P, whereas no differences were found between groups T and PT, suggesting a microbial evolution of the composition of group P towards group T over chronological age. Full-term delivery was associated with a greater diversity of microbes in HM. The microbial composition of pre-term HM, at the corrected age, did not show significant differences, as compared to the samples obtained from the full-term group, suggesting that it would be appropriate to consider the corrected age in terms of the composition and the diversity of the milk in future studies.
Collapse
Affiliation(s)
- Sofía Oddi
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Anastasia Mantziari
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Paula Huber
- Laboratorio de Plancton, Instituto Nacional de Limnología (INALI, UNL-CONICET), Universidad Nacional del Litoral, Santa Fe 3000, Argentina
- Departamento de Hydrobiologia, Universidade Federal de São Carlos (UFSCar), Rodovia Washington Luiz, São Carlos 13565-905, SP, Brazil
| | - Ana Binetti
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (INLAIN, UNL-CONICET), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe 3000, Argentina
| |
Collapse
|
126
|
Smilowitz JT, Allen LH, Dallas DC, McManaman J, Raiten DJ, Rozga M, Sela DA, Seppo A, Williams JE, Young BE, McGuire MK. Ecologies, synergies, and biological systems shaping human milk composition-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 2. Am J Clin Nutr 2023; 117 Suppl 1:S28-S42. [PMID: 37173059 DOI: 10.1016/j.ajcnut.2022.11.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 05/15/2023] Open
Abstract
Human milk is universally recognized as the preferred food for infants during the first 6 mo of life because it provides not only essential and conditionally essential nutrients in necessary amounts but also other biologically active components that are instrumental in protecting, communicating important information to support, and promoting optimal development and growth in infants. Despite decades of research, however, the multifaceted impacts of human milk consumption on infant health are far from understood on a biological or physiological basis. Reasons for this lack of comprehensive knowledge of human milk functions are numerous, including the fact that milk components tend to be studied in isolation, although there is reason to believe that they interact. In addition, milk composition can vary greatly within an individual as well as within and among populations. The objective of this working group within the Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN) Project was to provide an overview of human milk composition, factors impacting its variation, and how its components may function to coordinately nourish, protect, and communicate complex information to the recipient infant. Moreover, we discuss the ways whereby milk components might interact such that the benefits of an intact milk matrix are greater than the sum of its parts. We then apply several examples to illustrate how milk is better thought of as a biological system rather than a more simplistic "mixture" of independent components to synergistically support optimal infant health.
Collapse
Affiliation(s)
- Jennifer T Smilowitz
- Department of Food Science and Technology, University of California Davis, Davis, CA, USA; Foods for Health Institute, University of California Davis, Davis, CA, USA.
| | - Lindsay H Allen
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, University of California Davis, Davis, CA, USA
| | - David C Dallas
- Nutrition Program, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | - James McManaman
- Division of Reproductive Sciences, University of Colorado, Aurora, CO, USA
| | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Mary Rozga
- Evidence Analysis Center, Academy of Nutrition and Dietetics, Chicago, IL, USA
| | - David A Sela
- Department of Food Science, University of Massachusetts, Amherst, MA, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Antti Seppo
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Janet E Williams
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, USA
| | - Bridget E Young
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, USA.
| |
Collapse
|
127
|
Sikder MAA, Rashid RB, Ahmed T, Sebina I, Howard DR, Ullah MA, Rahman MM, Lynch JP, Curren B, Werder RB, Simpson J, Bissell A, Morrison M, Walpole C, Radford KJ, Kumar V, Woodruff TM, Ying TH, Ali A, Kaiko GE, Upham JW, Hoelzle RD, Cuív PÓ, Holt PG, Dennis PG, Phipps S. Maternal diet modulates the infant microbiome and intestinal Flt3L necessary for dendritic cell development and immunity to respiratory infection. Immunity 2023; 56:1098-1114.e10. [PMID: 37003256 DOI: 10.1016/j.immuni.2023.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/28/2022] [Accepted: 03/02/2023] [Indexed: 04/03/2023]
Abstract
Poor maternal diet during pregnancy is a risk factor for severe lower respiratory infections (sLRIs) in the offspring, but the underlying mechanisms remain elusive. Here, we demonstrate that in mice a maternal low-fiber diet (LFD) led to enhanced LRI severity in infants because of delayed plasmacytoid dendritic cell (pDC) recruitment and perturbation of regulatory T cell expansion in the lungs. LFD altered the composition of the maternal milk microbiome and assembling infant gut microbiome. These microbial changes reduced the secretion of the DC growth factor Flt3L by neonatal intestinal epithelial cells and impaired downstream pDC hematopoiesis. Therapy with a propionate-producing bacteria isolated from the milk of high-fiber diet-fed mothers, or supplementation with propionate, conferred protection against sLRI by restoring gut Flt3L expression and pDC hematopoiesis. Our findings identify a microbiome-dependent Flt3L axis in the gut that promotes pDC hematopoiesis in early life and confers disease resistance against sLRIs.
Collapse
Affiliation(s)
- Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jason P Lynch
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Rhiannon B Werder
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Mark Morrison
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Brisbane, QLD 4102, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Carina Walpole
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia
| | - Kristen J Radford
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Tan Hui Ying
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ayesha Ali
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Gerard E Kaiko
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - John W Upham
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, Brisbane, QLD 4102, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia; Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Robert D Hoelzle
- The School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Páraic Ó Cuív
- Mater Research Institute, The University of Queensland, Translational Research Institute, Wolloongabba, Brisbane, QLD 4102, Australia; Microba Life Sciences, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Paul G Dennis
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia; The School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
128
|
Chen J, Li H, Zhao T, Chen K, Chen MH, Sun Z, Xu W, Maas K, Lester BM, Cong XS. The Impact of Early Life Experiences and Gut Microbiota on Neurobehavioral Development in Preterm Infants: A Longitudinal Cohort Study. Microorganisms 2023; 11:microorganisms11030814. [PMID: 36985387 PMCID: PMC10056840 DOI: 10.3390/microorganisms11030814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/11/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVES The objective of this study is to investigate the impact of early life experiences and gut microbiota on neurobehavioral development in preterm infants during neonatal intensive care unit (NICU) hospitalization. METHODS Preterm infants were followed from NICU admission until their 28th postnatal day or until discharge. Daily stool samples, painful/stressful experiences, feeding patterns, and other clinical and demographic data were collected. Gut microbiota was profiled using 16S rRNA sequencing, and operational taxonomic units (OTUs) were selected to predict the neurobehaviors. The neurobehavioral development was assessed by the Neonatal Neurobehavioral Scale (NNNS) at 36 to 38 weeks of post-menstrual age (PMA). Fifty-five infants who had NNNS measurements were included in the sparse log-contrast regression analysis. RESULTS Preterm infants who experienced a high level of pain/stress during the NICU hospitalization had higher NNNS stress/abstinence scores. Eight operational taxonomic units (OTUs) were identified to be associated with NNNS subscales after controlling demographic and clinical features, feeding patterns, and painful/stressful experiences. These OTUs and taxa belonging to seven genera, i.e., Enterobacteriaceae_unclassified, Escherichia-Shigella, Incertae_Sedis, Veillonella, Enterococcus, Clostridium_sensu_stricto_1, and Streptococcus with five belonging to Firmicutes and two belonging to Proteobacteria phylum. The enriched abundance of Enterobacteriaceae_unclassified (OTU17) and Streptococcus (OTU28) were consistently associated with less optimal neurobehavioral outcomes. The other six OTUs were also associated with infant neurobehavioral responses depending on days at NICU stay. CONCLUSIONS This study explored the dynamic impact of specific OTUs on neurobehavioral development in preterm infants after controlling for early life experiences, i.e., acute and chronic pain/stress and feeding in the NICU. The gut microbiota and acute pain/stressful experiences dynamically impact the neurobehavioral development in preterm infants during their NICU hospitalization.
Collapse
Affiliation(s)
- Jie Chen
- College of Nursing, Florida State University, Tallahassee, FL 32306, USA
- School of Nursing, University of Connecticut, Storrs, CT 06269, USA
| | - Hongfei Li
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA
| | - Tingting Zhao
- School of Nursing, University of Connecticut, Storrs, CT 06269, USA
- School of Nursing, Yale University, Orange, CT 06477, USA
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA
| | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA
| | - Zhe Sun
- Department of Statistics, University of Connecticut, Storrs, CT 06269, USA
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT 06520, USA
| | - Wanli Xu
- School of Nursing, University of Connecticut, Storrs, CT 06269, USA
| | - Kendra Maas
- Microbial Analysis, Resources, and Services (MARS), University of Connecticut, Storrs, CT 06269, USA
| | - Barry M Lester
- Brown Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Xiaomei S Cong
- School of Nursing, University of Connecticut, Storrs, CT 06269, USA
- School of Nursing, Yale University, Orange, CT 06477, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
129
|
Radford-Smith DE, Anthony DC. Mechanisms of Maternal Diet-Induced Obesity Affecting the Offspring Brain and Development of Affective Disorders. Metabolites 2023; 13:455. [PMID: 36984895 PMCID: PMC10053489 DOI: 10.3390/metabo13030455] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Depression and metabolic disease are common disorders that share a bidirectional relationship and continue to increase in prevalence. Maternal diet and maternal behaviour both profoundly influence the developmental trajectory of offspring during the perinatal period. At an epidemiological level, both maternal depression and obesity during pregnancy have been shown to increase the risk of neuropsychiatric disease in the subsequent generation. Considerable progress has been made to understand the mechanisms by which maternal obesity disrupts the developing offspring gut-brain axis, priming offspring for the development of affective disorders. This review outlines such mechanisms in detail, including altered maternal care, the maternal microbiome, inflammation, breast milk composition, and maternal and placental metabolites. Subsequently, offspring may be prone to developing gut-brain interaction disorders with concomitant changes to brain energy metabolism, neurotransmission, and behaviour, alongside gut dysbiosis. The gut microbiome may act as a key modifiable, and therefore treatable, feature of the relationship between maternal obesity and the offspring brain function. Further studies examining the relationship between maternal nutrition, the maternal microbiome and metabolites, and offspring neurodevelopment are warranted to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Daniel E. Radford-Smith
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX37JX, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX13TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| | - Daniel C. Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| |
Collapse
|
130
|
Chandran D, Warren K, McKeone D, Hicks SD. The Association between Infant Colic and the Multi-Omic Composition of Human Milk. Biomolecules 2023; 13:biom13030559. [PMID: 36979494 PMCID: PMC10046316 DOI: 10.3390/biom13030559] [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: 02/24/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Infant colic is a common condition with unclear biologic underpinnings and limited treatment options. We hypothesized that complex molecular networks within human milk (i.e., microbes, micro-ribonucleic acids (miRNAs), cytokines) would contribute to colic risk, while controlling for medical, social, and nutritional variables. This hypothesis was tested in a cohort of 182 breastfed infants, assessed with a modified Infant Colic Scale at 1 month. RNA sequencing was used to interrogate microbial and miRNA features. Luminex assays were used to measure growth factors and cytokines. Milk from mothers of infants with colic (n = 28) displayed higher levels of Staphylococcus (adj. p = 0.038, d = 0.30), miR-224-3p (adj. p = 0.023, d = 0.33), miR-125b-5p (adj. p = 0.028, d = 0.29), let-7a-5p (adj. p = 0.028, d = 0.27), and miR-205-5p (adj. p = 0.029, d = 0.26) compared to milk from non-colic mother-infant dyads (n = 154). Colic symptom severity was directly associated with milk hepatocyte growth factor levels (R = 0.21, p = 0.025). A regression model involving let-7a-5p, miR-29a-3p, and Lactobacillus accurately modeled colic risk (X2 = 16.7, p = 0.001). Molecular factors within human milk may impact colic risk, and provide support for a dysbiotic/inflammatory model of colic pathophysiology.
Collapse
Affiliation(s)
- Desirae Chandran
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Kaitlyn Warren
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Daniel McKeone
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Steven D Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
131
|
Kapoor B, Gulati M, Gupta R, Singla RK. Microbiota dysbiosis and myasthenia gravis: Do all roads lead to Rome? Autoimmun Rev 2023; 22:103313. [PMID: 36918089 DOI: 10.1016/j.autrev.2023.103313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Dysregulated immune system with a failure to recognize self from non-self-antigens is one of the common pathogeneses seen in autoimmune diseases. The complex interplay of genetic and environmental factors is important for the occurrence and development of the disease. Among the environmental factors, disturbed gut microbiota (gut dysbiosis) has recently attracted particular attention, especially with advancement in human microbiome research. Although the alterations in microbiota have been seen in various autoimmune diseases, including those of nervous system, there is paucity of information on neuromuscular system diseases. Myasthenia gravis (MG) is one such rare autoimmune disease of neuromuscular junction, and is caused by generation of pathogenic autoantibodies to components of the postsynaptic muscle endplate. In the recent years, accumulating evidences have endorsed the key role of host microbiota, particularly those of gut, in the pathogenesis of MG. Differential microbiota composition, characterized by increased abundance of Fusobacteria, Bacteroidetes, and Proteobacteria, and decreased abundance of Actinobacteria and Firmicutes, has been seen in MG patients in comparison to healthy subjects. Disturbance of microbiota composition, particularly reduced ratio of Firmicutes/Bacteroidetes, alter the gut permeability, subsequently triggering the immunological response. Resultant reduction in levels of short chain fatty acids (SCFAs) is another factor contributing to the immunological response in MG patients. Modulation of gut microbiota via intervention of probiotics, prebiotics, synbiotics, postbiotics (metabiotics), and fecal microbiota transplantation (FMT) is considered to be the futuristic approach for the management of MG. This review summarizes the role of gut microbiota and their metabolites (postbiotics) in the progression of MG. Also, various bacteriotherapeutic approaches involving gut microbiota are discussed for the prevention of MG progression.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia.
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Xinchuan Road, 2222, Chengdu, Sichuan, China; iGlobal Research and Publishing Foundation, New Delhi, India
| |
Collapse
|
132
|
Bogaert D, van Beveren GJ, de Koff EM, Lusarreta Parga P, Balcazar Lopez CE, Koppensteiner L, Clerc M, Hasrat R, Arp K, Chu MLJN, de Groot PCM, Sanders EAM, van Houten MA, de Steenhuijsen Piters WAA. Mother-to-infant microbiota transmission and infant microbiota development across multiple body sites. Cell Host Microbe 2023; 31:447-460.e6. [PMID: 36893737 DOI: 10.1016/j.chom.2023.01.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/07/2022] [Accepted: 01/30/2023] [Indexed: 03/11/2023]
Abstract
Early-life microbiota seeding and subsequent development is crucial to future health. Cesarean-section (CS) birth, as opposed to vaginal delivery, affects early mother-to-infant transmission of microbes. Here, we assess mother-to-infant microbiota seeding and early-life microbiota development across six maternal and four infant niches over the first 30 days of life in 120 mother-infant pairs. Across all infants, we estimate that on average 58.5% of the infant microbiota composition can be attributed to any of the maternal source communities. All maternal source communities seed multiple infant niches. We identify shared and niche-specific host/environmental factors shaping the infant microbiota. In CS-born infants, we report reduced seeding of infant fecal microbiota by maternal fecal microbes, whereas colonization with breastmilk microbiota is increased when compared with vaginally born infants. Therefore, our data suggest auxiliary routes of mother-to-infant microbial seeding, which may compensate for one another, ensuring that essential microbes/microbial functions are transferred irrespective of disrupted transmission routes.
Collapse
Affiliation(s)
- Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Gina J van Beveren
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | - Emma M de Koff
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | - Paula Lusarreta Parga
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Carlos E Balcazar Lopez
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Melanie Clerc
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Raiza Hasrat
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | - Kayleigh Arp
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | - Mei Ling J N Chu
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | - Pieter C M de Groot
- Department of Obstetrics and Gynaecology, Spaarne Gasthuis, 2035 RC Haarlem, the Netherlands
| | - Elisabeth A M Sanders
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands
| | | | - Wouter A A de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands; Centre for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, the Netherlands.
| |
Collapse
|
133
|
Guo W, Bi SS, Wang WW, Zhou M, Neves ALA, Degen AA, Guan LL, Long RJ. Maternal rumen and milk microbiota shape the establishment of early-life rumen microbiota in grazing yak calves. J Dairy Sci 2023; 106:2054-2070. [PMID: 36710176 DOI: 10.3168/jds.2022-22655] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/05/2022] [Indexed: 01/30/2023]
Abstract
Early-life gut microbial colonization and development exert a profound impact on the health and metabolism of the host throughout the life span. The transmission of microbes from the mother to the offspring affects the succession and establishment of the early-life rumen microbiome in newborns, but the contributions of different maternal sites to the rumen microbial establishment remain unclear. In the present study, samples from different dam sites (namely, oral, rumen fluid, milk, and teat skin) and rumen fluid of yak calves were collected at 6 time points between d 7 and 180 postpartum to determine the contributions of the different maternal sites to the establishment of the bacterial and archaeal communities in the rumen during early life. Our analysis demonstrated that the dam's microbial communities clustered according to the sites, and the calves' rumen microbiota resembled that of the dam consistently regardless of fluctuations at d 7 and 14. The dam's rumen microbiota was the major source of the calves' rumen bacteria (7.9%) and archaea (49.7%) compared with the other sites, whereas the potential sources of the calf rumen microbiota from other sites varied according to the age. The contribution of dam's rumen bacteria increased with age from 0.36% at d 7 to 14.8% at d 180, whereas the contribution of the milk microbiota showed the opposite trend, with its contribution reduced from 2.7% at d 7 to 0.2% at d 180. Maternal oral archaea were the main sources of the calves' rumen archaea at d 14 (50.4%), but maternal rumen archaea became the main source gradually and reached 66.2% at d 180. These findings demonstrated the potential microbial transfer from the dam to the offspring that could influence the rumen microbiota colonization and establishment in yak calves raised under grazing regimens, providing the basis for future microbiota manipulation strategies during their early life.
Collapse
Affiliation(s)
- W Guo
- State Key Laboratory of Grassland Agro-ecosystems, International Centre of Tibetan Plateau Ecosystem Management, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - S S Bi
- State Key Laboratory of Grassland Agro-ecosystems, International Centre of Tibetan Plateau Ecosystem Management, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - W W Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang 550025, China
| | - M Zhou
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - A L A Neves
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 3, DK-1870, Frederiksberg C, Denmark
| | - A A Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva 8410500, Israel
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada.
| | - R J Long
- State Key Laboratory of Grassland Agro-ecosystems, International Centre of Tibetan Plateau Ecosystem Management, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
134
|
Wu S, Ren L, Li J, Shen X, Zhou Q, Miao Z, Jia W, He F, Cheng R. Breastfeeding might partially contribute to gut microbiota construction and stabilization of propionate metabolism in cesarean-section infants. Eur J Nutr 2023; 62:615-631. [PMID: 36173468 DOI: 10.1007/s00394-022-03020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE This study was aimed to determine how delivery mode and feeding pattern influence the infant's gut microbiota construction and the variation of fecal microbial metabolites from a birth cohort. METHODS Fecal samples collected from 61 full-term born Chinese infants at four time points: day 0, day 7, month 1, and month 3. Based on delivery mode (vaginal delivery [V] or cesarean section [C]) and feeding pattern (breastfeeding [B] or mixed feeding [M]), infants were divided into four groups, namely VB, CB, VM, and CM groups. The gut microbiota composition and bacterial diversity were assessed using 16S rRNA sequencing. Short-chain fatty acid (SCFA) concentrations were determined via gas chromatography-mass spectrometry (GC-MS). RESULTS The CM group had a significantly higher relative abundance of Firmicutes (day 0 and month 1), Enterococcaceae (month 3), and Enterococcus (month 3) than the VB group and a significantly higher abundance of Firmicutes (month 1) and Blautia (month 3) than the CB group. The VB and CB groups exhibited a stable SCFA variation and a significantly lower level of propionate compared with the VM and CM groups. All groups showed an intense transition of enterotypes within 1 month and became stable at 3 months. The correlation between SCFA and enterotypes showed a significant positive correlation between Bifidobacteriaceae and acetate in the CB group (day 7 and month 3) and a significant positive correlation between Clostridiaceae and butyrate in the CB and VB groups (day 7 and month 3), respectively. CONCLUSION These results indicated that C-section was associated with higher abundance of the phylum Firmicutes and family Enterococcaceae, and intense fluctuation of SCFA, at least propionate. And breastfeeding might partially contribute to gut microbiota construction and stabilization propionate metabolism in cesarean-section infants.
Collapse
Affiliation(s)
- Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lei Ren
- Hebei Inatural Bio-Tech Co.,Ltd., Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qingqing Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhonghua Miao
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wen Jia
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
135
|
Newman TM, Clear KYJ, Wilson AS, Soto-Pantoja DR, Ochs-Balcom HM, Cook KL. Early-life dietary exposures mediate persistent shifts in the gut microbiome and visceral fat metabolism. Am J Physiol Cell Physiol 2023; 324:C644-C657. [PMID: 35848617 PMCID: PMC9970661 DOI: 10.1152/ajpcell.00380.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In utero dietary exposures are linked to the development of metabolic syndrome in adult offspring. These dietary exposures can potentially impact gut microbial composition and offspring metabolic health. Female BALB/c mice were administered a lard, lard + flaxseed oil, high sugar, or control diet 4 wk before mating, throughout mating, pregnancy, and lactation. Female offspring were offered low-fat control diet at weaning. Fecal 16S sequencing was performed. Untargeted metabolomics was performed on visceral adipose tissue (VAT) of adult female offspring. Immunohistochemistry was used to determine adipocyte size, VAT collagen deposition, and macrophage content. Hippurate was administered via weekly intraperitoneal injections to low-fat and high-fat diet-fed female mice and VAT fibrosis and collagen 1A (COL1A) were assessed by immunohistochemistry. Lard diet exposure was associated with elevated body and VAT weight and dysregulated glucose metabolism. Lard + flaxseed oil attenuated these effects. Lard diet exposures were associated with increased adipocyte diameter and VAT macrophage count. Lard + flaxseed oil reduced adipocyte diameter and fibrosis compared with the lard diet. Hippurate-associated bacteria were influenced by lard versus lard + flax exposures that persisted to adulthood. VAT hippurate was increased in lard + flaxseed oil compared with lard diet. Hippurate supplementation mitigated VAT fibrosis pathology. Maternal high-fat lard diet consumption resulted in long-term metabolic and gut microbiome programming in offspring, impacting VAT inflammation and fibrosis, and was associated with reduced VAT hippurate content. These traits were not observed in maternal high-fat lard + flaxseed oil diet-exposed offspring. Hippurate supplementation reduced VAT fibrosis. These data suggest that detrimental effects of early-life high-fat lard diet exposure can be attenuated by dietary omega-3 polyunsaturated fatty acid supplementation.
Collapse
Affiliation(s)
- Tiffany M. Newman
- 1Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina,2Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kenysha Y. J. Clear
- 2Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Adam S. Wilson
- 2Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - David R. Soto-Pantoja
- 1Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina,2Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina,3Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Heather M. Ochs-Balcom
- 4Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Katherine L. Cook
- 1Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina,2Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina,3Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
136
|
Korobkova L, Morin EL, Aoued H, Sannigrahi S, Garza KM, Siebert ER, Walum H, Cabeen RP, Sanchez MM, Dias BG. RNA in extracellular vesicles during adolescence reveal immune, energetic and microbial imprints of early life adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529808. [PMID: 36865138 PMCID: PMC9980043 DOI: 10.1101/2023.02.23.529808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Exposure to early life adversity (ELA), including childhood maltreatment, is one of the most significant risk factors for the emergence of neuropsychiatric disorders in adolescence and adulthood. Despite this relationship being well established, the underlying mechanisms remain unclear. One way to achieve this understanding is to identify molecular pathways and processes that are perturbed as a consequence of childhood maltreatment. Ideally, these perturbations would be evident as changes in DNA, RNA or protein profiles in easily accessible biological samples collected in the shadow of childhood maltreatment. In this study, we isolated circulating extracellular vesicles (EVs) from plasma collected from adolescent rhesus macaques that had either experienced nurturing maternal care (CONT) or maternal maltreatment (MALT) in infancy. RNA sequencing of RNA in plasma EVs and gene enrichment analysis revealed that genes related to translation, ATP synthesis, mitochondrial function and immune response were downregulated in MALT samples, while genes involved in ion transport, metabolism and cell differentiation were upregulated. Interestingly, we found that a significant proportion of EV RNA aligned to the microbiome and that MALT altered the diversity of microbiome-associated RNA signatures found in EVs. Part of this altered diversity suggested differences in prevalence of bacterial species in CONT and MALT animals noted in the RNA signatures of the circulating EVs. Our findings provide evidence that immune function, cellular energetics and the microbiome may be important conduits via which infant maltreatment exerts effects on physiology and behavior in adolescence and adulthood. As a corollary, perturbations of RNA profiles related to immune function, cellular energetics and the microbiome may serve as biomarkers of responsiveness to ELA. Our results demonstrate that RNA profiles in EVs can serve as a powerful proxy to identify biological processes that might be perturbed by ELA and that may contribute to the etiology of neuropsychiatric disorders in the aftermath of ELA.
Collapse
|
137
|
Natural Green Spaces, Sensitization to Allergens, and the Role of Gut Microbiota during Infancy. mSystems 2023; 8:e0119022. [PMID: 36790181 PMCID: PMC10134798 DOI: 10.1128/msystems.01190-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
The environment plays an instrumental role in the developmental origins of health and disease. Protective features of the environment in the development of asthma and atopy have been insufficiently studied. We used data from the CHILD (Canadian Healthy Infant Longitudinal Development) Cohort Study to examine relationships between living near natural green spaces in early infancy in Edmonton, AB, Canada and the development of atopic sensitization at 1 year and 3 years of age in a cohort of 699 infants, and whether these associations were mediated by infant gut microbiota (measured using 16s V4 amplicon sequencing) at 4 months. The Urban Planning Land Vegetation Index (uPLVI) map of the City of Edmonton was used to assess infants' exposure to natural spaces based on their home postal codes, and atopic sensitization was assessed using skin prink testing (SPTs) for common food and inhalant allergens. Our findings suggest there is a protective effect of natural green space proximity on the development of multiple inhalant atopic sensitizations at 3 years (odds ratio = 0.28 [95% CI 0.09, 0.90]). This relationship was mediated by changes to Actinobacteria diversity in infant fecal samples taken at 4 months. We also found a positive association between nature proximity and sensitization to at least one food or inhaled allergen; this association was not mediated by gut microbiota. Together, these findings underscore the importance of promoting natural urban greenspace preservation to improve child health by reducing atopic disease susceptibility. IMPORTANCE Our findings highlight the importance of preserving natural green space in urban settings to prevent sensitization to environmental allergens and promote early-life gut microbiota pathways to this health benefit. These findings support a mediating role of gut microbiome compositions in health and disease susceptibility. This study used unique, accurate, and comprehensive methodology to classify natural space exposure via a high-resolution topographical map of foliage subtypes within the City of Edmonton limits. These methods are improvements from other methods previously used to classify natural space exposure, such as the normalized density vegetation index from satellite imagery, which is not able to distinguish anthropogenic from green space. The use of these methods and the associations found between natural green space exposure and atopic sensitization outcomes support their use in future studies. Our findings also provide many avenues for future research including longer term follow up of this cohort and investigation of a causal role of reduced Actinobacteria diversity on atopic sensitization development.
Collapse
|
138
|
Olga L, van Diepen JA, Chichlowski M, Petry CJ, Vervoort J, Dunger DB, Kortman GAM, Gross G, Ong KK. Butyrate in Human Milk: Associations with Milk Microbiota, Milk Intake Volume, and Infant Growth. Nutrients 2023; 15:916. [PMID: 36839274 PMCID: PMC9963357 DOI: 10.3390/nu15040916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
Butyrate in human milk (HM) has been suggested to reduce excessive weight and adipo-sity gains during infancy. However, HM butyrate's origins, determinants, and its influencing mechanism on weight gain are not completely understood. These were studied in the prospective longitudinal Cambridge Baby Growth and Breastfeeding Study (CBGS-BF), in which infants (n = 59) were exclusively breastfed for at least 6 weeks. Infant growth (birth, 2 weeks, 6 weeks, 3 months, 6 months, and 12 months) and HM butyrate concentrations (2 weeks, 6 weeks, 3 months, and 6 months) were measured. At age 6 weeks, HM intake volume was measured by deuterium-labelled water technique and HM microbiota by 16S sequencing. Cross-sectionally at 6 weeks, HM butyrate was associated with HM microbiota composition (p = 0.036) although no association with the abundance of typical butyrate producers was detected. In longitudinal analyses across all time points, HM butyrate concentrations were overall negatively associated with infant weight and adiposity, and associations were stronger at younger infant ages. HM butyrate concentration was also inversely correlated with HM intake volume, supporting a possible mechanism whereby butyrate might reduce infant growth via appetite regulation and modulation of HM intake.
Collapse
Affiliation(s)
- Laurentya Olga
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Janna A van Diepen
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN 47721, USA
| | - Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN 47721, USA
| | - Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jacques Vervoort
- Department of Agrotechnology and Food Sciences, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0SL, UK
| | | | - Gabriele Gross
- Medical and Scientific Affairs, Reckitt/Mead Johnson Nutrition Institute, Evansville, IN 47721, USA
| | - Ken K Ong
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
- MRC Epidemiology Unit, Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0SL, UK
- Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
139
|
Liu S, Zhang Z, Ma L. A Review Focusing on Microbial Vertical Transmission during Sow Pregnancy. Vet Sci 2023; 10:vetsci10020123. [PMID: 36851427 PMCID: PMC9967962 DOI: 10.3390/vetsci10020123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Microorganisms are closely related to the body's physiological activities and growth and development of the body, and participate in many physiological metabolic activities. Analysis of the structure and source of early colonizing bacteria in the intestinal tract of humans and rodents shows that early colonizing bacteria in the intestinal tract of mammals have solid maternal characteristics, and maternal microbes play an essential role in the formation of progeny intestinal flora. The placental microbiome, maternal microbiome and breast milk microbiome are currently hot topics in the field of life science. This paper discusses the vertical transmission and endogenous sources of the mother-to-piglet microbiome through these three pathways, aiming to provide a new research idea for intervention in the intestinal microbiome in young piglets.
Collapse
Affiliation(s)
- Shengjun Liu
- Jiangxi Haida Feed Co., Ltd., Nanchang 331700, China
| | - Zixi Zhang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Longteng Ma
- Jiangxi Haida Feed Co., Ltd., Nanchang 331700, China
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Correspondence:
| |
Collapse
|
140
|
Ruiz-Ojeda FJ, Plaza-Diaz J, Morales J, Álvarez-Calatayud G, Climent E, Silva Á, Martinez-Blanch JF, Enrique M, Tortajada M, Ramon D, Alvarez B, Chenoll E, Gil Á. Effects of a Novel Infant Formula on the Fecal Microbiota in the First Six Months of Life: The INNOVA 2020 Study. Int J Mol Sci 2023; 24:3034. [PMID: 36769356 PMCID: PMC9917896 DOI: 10.3390/ijms24033034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Exclusive breastfeeding is highly recommended for infants for at least the first six months of life. However, for some mothers, it may be difficult or even impossible to do so. This can lead to disturbances in the gut microbiota, which in turn may be related to a higher incidence of acute infectious diseases. Here, we aimed to evaluate whether a novel starting formula versus a standard formula provides a gut microbiota composition more similar to that of breastfed infants in the first 6 months of life. Two hundred and ten infants (70/group) were enrolled in the study and completed the intervention until 12 months of age. For the intervention period, infants were divided into three groups: Group 1 received formula 1 (INN) with a lower amount of protein, a proportion of casein to whey protein ratio of about 70/30 by increasing the content of α-lactalbumin, and with double the amount of docosahexaenoic acid/arachidonic acid than the standard formula; INN also contained a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis). Group 2 received the standard formula (STD) and the third group was exclusively breastfed (BF) for exploratory analysis. During the study, visits were made at 21 days, 2, 4, and 6 months of age, with ±3 days for the visit at 21 days of age, ±1 week for the visit at 2 months, and ±2 weeks for the others. Here, we reveal how consuming the INN formula promotes a similar gut microbiota composition to those infants that were breastfed in terms of richness and diversity, genera, such as Bacteroides, Bifidobacterium, Clostridium, and Lactobacillus, and calprotectin and short-chain fatty acid levels at 21 days, 2 and 6 months. Furthermore, we observed that the major bacteria metabolic pathways were more alike between the INN formula and BF groups compared to the STD formula group. Therefore, we assume that consumption of the novel INN formula might improve gut microbiota composition, promoting a healthier intestinal microbiota more similar to that of an infant who receives exclusively human milk.
Collapse
Affiliation(s)
- Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Neuherberg, 85764 Munich, Germany
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Spain
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Javier Morales
- Product Development Department, Alter Farmacia SA, 28880 Madrid, Spain
| | | | - Eric Climent
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Ángela Silva
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | | | - María Enrique
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Marta Tortajada
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Daniel Ramon
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Beatriz Alvarez
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Empar Chenoll
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Spain
- CIBEROBN, CIBER Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
141
|
Couvillion SP, Mostoller KE, Williams JE, Pace RM, Stohel IL, Peterson HK, Nicora CD, Nakayasu ES, Webb-Robertson BJM, McGuire MA, McGuire MK, Metz TO. Interrogating the role of the milk microbiome in mastitis in the multi-omics era. Front Microbiol 2023; 14:1105675. [PMID: 36819069 PMCID: PMC9932517 DOI: 10.3389/fmicb.2023.1105675] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
There is growing interest in a functional understanding of milk-associated microbiota as there is ample evidence that host-associated microbial communities play an active role in host health and phenotype. Mastitis, characterized by painful inflammation of the mammary gland, is prevalent among lactating humans and agricultural animals and is associated with significant clinical and economic consequences. The etiology of mastitis is complex and polymicrobial and correlative studies have indicated alterations in milk microbial community composition. Recent evidence is beginning to suggest that a causal relationship may exist between the milk microbiota and host phenotype in mastitis. Multi-omic approaches can be leveraged to gain a mechanistic, molecular level understanding of how the milk microbiome might modulate host physiology, thereby informing strategies to prevent and ameliorate mastitis. In this paper, we review existing studies that have utilized omics approaches to investigate the role of the milk microbiome in mastitis. We also summarize the strengths and challenges associated with the different omics techniques including metagenomics, metatranscriptomics, metaproteomics, metabolomics and lipidomics and provide perspective on the integration of multiple omics technologies for a better functional understanding of the milk microbiome.
Collapse
Affiliation(s)
- Sneha P. Couvillion
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,*Correspondence: Sneha P. Couvillion, ✉
| | - Katie E. Mostoller
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Janet E. Williams
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Ryan M. Pace
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Izabel L. Stohel
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Haley K. Peterson
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Carrie D. Nicora
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Ernesto S. Nakayasu
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Bobbie-Jo M. Webb-Robertson
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States
| | - Mark A. McGuire
- Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, United States
| | - Michelle K. McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, United States
| | - Thomas O. Metz
- Pacific Northwest National Laboratory, Earth and Biological Sciences Directorate, Richland, WA, United States,Thomas O. Metz, ✉
| |
Collapse
|
142
|
Lif Holgerson P, Esberg A, West CE, Johansson I. The breast milk and childhood gastrointestinal microbiotas and disease outcomes: a longitudinal study. Pediatr Res 2023; 93:570-578. [PMID: 36216869 PMCID: PMC9988688 DOI: 10.1038/s41390-022-02328-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND We aimed to characterize breast milk microbiota and define associations with saliva and fecal microbiota and selected diseases in preschool children. METHODS In a longitudinal cohort study, the microbiotas from breast milk, mouth, and fecal samples were characterized by 16S rRNA gene sequencing. Questionnaires and medical records provided information on demographics, medical, and dental data. RESULTS The phylogeny in breast milk, saliva swabs, and feces differed at all levels (p < 0.0003), though all harbored species in Streptococcus, Veillonella, and Haemophilus. Species richness was highest in breast milk with increasing resemblance with the oral swab microbiota by increasing age. Caries-affected children at age 5 had been fed breast milk with tenfold higher abundance of caries-associated bacteria, e.g., Streptococcus mutans, than caries-free children (p < 0.002). At that age, taxa, e.g., Neisseria sicca were overrepresented in saliva swabs of children with otitis media (LDA score >2, p < 0.05). Gut symbionts, e.g., Bacteroides, were underrepresented in 3-month fecal samples in children later diagnosed with allergic disease (LDA score >2, p < 0.05). CONCLUSIONS Distinct microbiotas for the three sources were confirmed, though resemblance between milk and oral swab microbiota increased by age. Future studies should evaluate if the observed associations with disease outcomes are causal. IMPACT Few studies have studied the association between breast milk microbiota and gastrointestinal microbiota beyond early infancy. The present study confirms distinct microbiota profiles in breast milk, saliva swabs, and feces in infancy and indicates increasing resemblance between breast milk and the oral microbiota by increasing age. The fecal microbiota at 3 months was associated with later allergic disease; the saliva microbiota by age 5 differed between children with and without otitis media at the same age; and children with caries by age 5 had been fed breast milk with a higher abundance of caries-associated bacteria.
Collapse
Affiliation(s)
- Pernilla Lif Holgerson
- Department of Odontology, Section of Pediatric Dentistry, Umeå University, Umeå, Sweden.
| | - Anders Esberg
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| | - Christina E West
- Department of Clinical Sciences, Section of Pediatrics, Umeå University, Umeå, Sweden
| | - Ingegerd Johansson
- Department of Odontology, Section of Cariology, Umeå University, Umeå, Sweden
| |
Collapse
|
143
|
Simon DA, Kellermayer R. Disturbed Pediatric Gut Microbiome Maturation in the Developmental Origins of Subsequent Chronic Disease. J Pediatr Gastroenterol Nutr 2023; 76:123-127. [PMID: 36705694 DOI: 10.1097/mpg.0000000000003664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The microbiome is known to play an important role in the development and maintenance of human health. During early childhood the gut microbiome undergoes a rapid evolution, making this developmental window most susceptible to microbial manipulation and, therefore, most vulnerable to environmental stimuli. Such stimuli may induce persistent alterations (or dysbiosis) in microbiome and/or host physiology, thereby resulting in susceptibility to subsequent disease development. This phenomenon is frequently described as "the microbial developmental origins of disease." In this topic of the month, we call attention to the microbial developmental origins of disease by examining the potential for childhood antibiotic exposures and appendectomy (ie, inducers of dysbiosis) to influence the pathogenesis of certain multifactorial, common diseases (eg, celiac disease, inflammatory bowel disease, obesity), especially those with increasing incidence worldwide. We conclude that fully appreciating the critical components in the microbial developmental origins of common chronic disorders is a major task ahead of pediatric gastroenterologists in the 21st century. Such information will be key in working to prevent numerous common and emerging disorders.
Collapse
Affiliation(s)
- David Aaron Simon
- From the Division of Pediatric Gastroenterology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
| | - Richard Kellermayer
- From the Division of Pediatric Gastroenterology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX
- the USDA/ARS Children's Nutrition Research Center, Houston, TX
| |
Collapse
|
144
|
Fenneman AC, Weidner M, Chen LA, Nieuwdorp M, Blaser MJ. Antibiotics in the pathogenesis of diabetes and inflammatory diseases of the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2023; 20:81-100. [PMID: 36258032 PMCID: PMC9898198 DOI: 10.1038/s41575-022-00685-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 02/06/2023]
Abstract
Antibiotic use is increasing worldwide. However, the use of antibiotics is clearly associated with changes in gut microbiome composition and function, and perturbations have been identified as potential environmental risk factors for chronic inflammatory disorders of the gastrointestinal tract. In this Review, we examine the association between the use of antibiotics and the onset and development of both type 1 and type 2 diabetes, inflammatory bowel disease, including ulcerative colitis and Crohn's disease, as well as coeliac disease and eosinophilic oesophagitis. We discuss the key findings of epidemiological studies, provide mechanistic insights into the pathways by which the gut microbiota might contribute to these diseases, and assess clinical trials investigating the effects of antibiotics. Such studies indicate that antibiotic exposures, varying in type, timing and dosage, could explain differences in disease risk. There seems to be a critical window in early life in which perturbation of the microbiome has a substantial effect on disease development. Identifying the antibiotic-perturbed gut microbiota as a factor that contributes to the pathophysiology of these inflammatory disorders might stimulate new approaches to prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Aline C Fenneman
- Department of Clinical and Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Melissa Weidner
- Department of Paediatrics, Rutgers University, New Brunswick, NJ, USA
| | - Lea Ann Chen
- Department of Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Max Nieuwdorp
- Department of Clinical and Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences (ACS), Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Martin J Blaser
- Department of Medicine, Rutgers University, New Brunswick, NJ, USA.
- Department of Pathology and Laboratory Medicine, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
145
|
Gao LM, Liu GY, Wang HL, Wassie T, Wu X, Yin YL. Impact of dietary supplementation with N-carbamoyl-aspartic acid on serum metabolites and intestinal microflora of sows. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:750-763. [PMID: 36054758 DOI: 10.1002/jsfa.12186] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/25/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND N-Carbamoyl-aspartic acid (NCA) is a critical precursor for de novo biosynthesis of pyrimidine nucleotides. To investigate the cumulative effects of maternal supplementation with NCA on the productive performance, serum metabolites and intestinal microbiota of sows, 40 pregnant sows (∼day 80) were assigned into two groups: (1) the control (CON) and (2) treatment (NCA, 50 g t-1 NCA). RESULTS Results showed that piglets from the NCA group had heavier birth weight than those in the CON group (P < 0.05). In addition, maternal supplementation with NCA decreased the backfat loss of sows during lactation (P < 0.05). Furthermore,16S-rRNA sequencing results revealed that maternal NCA supplementation decreased the abundance of Cellulosilyticum, Fournierella, Anaerovibrio, and Oribacterium genera of sows during late pregnancy (P < 0.05). Similarly, on the 14th day of lactation, maternal supplementation with NCA reduced the diversity of fecal microbes of sows as evidenced by significantly lower observed species, Chao1, and Ace indexes, and decreased the abundance of Lachnospire, Faecalibacterium, and Anaerovorax genera, while enriched the abundance of Catenisphaera (P < 0.05). Untargeted metabolomics showed that a total of 48 differentially abundant biomarkers were identified, which were mainly involved in metabolic pathways of arginine/proline metabolism, phenylalanine/tyrosine metabolism, and fatty acid biosynthesis, etc. CONCLUSION: Overall, the results indicated that NCA supplementation regulated intestinal microbial composition of sows and serum differential metabolites related to arginine, proline, phenylalanine, tyrosine, and fatty acids metabolism that may contribute to regulating the backfat loss of sows, and the birth weight and diarrhea rate of piglets. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lu-Min Gao
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, China
| | - Gang-Yi Liu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
| | - Hong-Ling Wang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
| | - Teketay Wassie
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
| | - Xin Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Yu-Long Yin
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Science, Beijing, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| |
Collapse
|
146
|
Johnson KE, Heisel T, Allert M, Fürst A, Yerabandi N, Knights D, Jacobs KM, Lock EF, Bode L, Fields DA, Rudolph MC, Gale CA, Albert FW, Demerath EW, Blekhman R. Human milk variation is shaped by maternal genetics and impacts the infant gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525211. [PMID: 36747843 PMCID: PMC9900818 DOI: 10.1101/2023.01.24.525211] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Human milk is a complex mix of nutritional and bioactive components that provide complete nutrition for the infant. However, we lack a systematic knowledge of the factors shaping milk composition and how milk variation influences infant health. Here, we used multi-omic profiling to characterize interactions between maternal genetics, milk gene expression, milk composition, and the infant fecal microbiome in 242 exclusively breastfeeding mother-infant pairs. We identified 487 genetic loci associated with milk gene expression unique to the lactating mammary gland, including loci that impacted breast cancer risk and human milk oligosaccharide concentration. Integrative analyses uncovered connections between milk gene expression and infant gut microbiome, including an association between the expression of inflammation-related genes with IL-6 concentration in milk and the abundance of Bifidobacteria in the infant gut. Our results show how an improved understanding of the genetics and genomics of human milk connects lactation biology with maternal and infant health.
Collapse
Affiliation(s)
- Kelsey E Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Timothy Heisel
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mattea Allert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Annalee Fürst
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Nikhila Yerabandi
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Dan Knights
- BioTechnology Institute, College of Biological Sciences, University of Minnesota, Minneapolis, MN, USA
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Katherine M Jacobs
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal-Fetal Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Eric F Lock
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Human Milk Institute (HMI) and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA
| | - David A Fields
- Department of Pediatrics, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, Department of Physiology, the University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Cheryl A Gale
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, USA
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Division of Biological Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
147
|
Gildner TE, Cepon-Robins TJ. Rural Embodiment and Community Health: an Anthropological Case Study on Biocultural Determinants of Tropical Disease Infection and Immune System Development in the USA. CURRENT TROPICAL MEDICINE REPORTS 2023; 10:26-39. [PMID: 36714157 PMCID: PMC9868515 DOI: 10.1007/s40475-023-00282-z] [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] [Accepted: 12/12/2022] [Indexed: 01/24/2023]
Abstract
Purpose of Review Biocultural methods are critically important for identifying environmental and socioeconomic factors linked with tropical disease risk and outcomes. For example, embodiment theory refers to the process by which lived experiences impact individual biology. Increased exposure to pathogens, chronic psychosocial stress, and unequal resource access are all outcomes linked with discrimination and poverty. Through lived experiences, race and socioeconomic inequality can literally become embodied-get under the skin and affect physiology-impacting immune responses and contributing to lifelong health disparities. Yet, few studies have investigated tropical disease patterns and associated immune function using embodiment theory to understand lasting physiological impacts associated with living in a high-pathogen environment. Recent Findings Here, we use preliminary data drawn from the Rural Embodiment and Community Health (REACH) study to assess whether pathogen exposure and immune stimulation within a sample of children from the Mississippi Delta are associated with household income. We also test whether immune marker levels-assessed with enzyme-linked immunosorbent assays using dried blood spot samples-vary between the REACH sample and a similarly aged nationally representative NHANES sample. Immune marker levels did not differ significantly between REACH participants living below vs. above the federal poverty line, yet immunoglobulin E levels-a marker of macroparasite infection-were higher among REACH study participants compared to the NHANES sample. Summary These results may suggest community-level pathogenic exposures (i.e., parasitic infections) are embodied by REACH participants with implications for long-term immune function, potentially resulting in immune aspects that differ from nationally representative samples. Supplementary Information The online version contains supplementary material available at 10.1007/s40475-023-00282-z.
Collapse
Affiliation(s)
- Theresa E Gildner
- Department of Anthropology, Washington University in St. Louis, St. Louis, MO USA
| | - Tara J Cepon-Robins
- Department of Anthropology, University of Colorado Colorado Springs, Colorado Springs, CO USA
| |
Collapse
|
148
|
Wang H, Zhou C, Gu S, Sun Y. Surrogate fostering of mice prevents prenatal estradiol-induced insulin resistance via modulation of the microbiota-gut-brain axis. Front Microbiol 2023; 13:1050352. [PMID: 36699605 PMCID: PMC9868306 DOI: 10.3389/fmicb.2022.1050352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Prenatal and early postnatal development are known to influence future health. We previously reported that prenatal high estradiol (HE) exposure induces insulin resistance in male mice by disrupting hypothalamus development. Because a foster dam can modify a pup's gut microbiota and affect its health later in life, we explored whether surrogate fostering could also influence glucose metabolism in HE offspring and examined mechanisms that might be involved. Methods We performed a surrogate fostering experiment in mice and examined the relationship between the metabolic markers associated to insulin resistance and the composition of the gut microbiota. Results HE pups raised by HE foster dams (HE-HE) developed insulin resistance, but HE pups fostered by negative control dams (NC-HE) did not. The gut microbiota composition of HE-HE mice differed from that of NC mice raised by NC foster dams (NC-NC), whereas the composition in NC-HE mice was similar to that of NC-NC mice. Compared with NC-NC mice, HE-HE mice had decreased levels of fecal short-chain fatty acids and serum intestinal hormones, increased food intake, and increased hypothalamic neuropeptide Y expression. In contrast, none of these indices differed between NC-HE and NC-NC mice. Spearman correlation analysis revealed a significant correlation between the altered gut microbiota composition and the insulin resistance-related metabolic indicators, indicating involvement of the microbiota-gut-brain axis. Discussion Our findings suggest that alterations in the early growth environment may prevent fetal-programmed glucose metabolic disorder via modulation of the microbiota-gut-brain axis. These findings offer direction for development of translational solutions for adult diseases associated with aberrant microbial communities in early life.
Collapse
Affiliation(s)
- Huihui Wang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China,Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chengliang Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Shuping Gu
- Department of Science and Technology Research, Shanghai Model Organisms, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China,Animal Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Yun Sun, ✉
| |
Collapse
|
149
|
Chen J, Li H, Zhao T, Chen K, Chen MH, Sun Z, Xu W, Maas K, Lester B, Cong X. The impact of early life experiences and gut microbiota on neurobehavioral development among preterm infants: A longitudinal cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.04.23284200. [PMID: 36711616 PMCID: PMC9882379 DOI: 10.1101/2023.01.04.23284200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objectives To investigate the impact of early life experiences and gut microbiota on neurobehavioral development among preterm infants during neonatal intensive care unit (NICU) hospitalization. Methods Preterm infants were followed from the NICU admission until their 28 th postnatal day or until discharge. Daily stool samples, painful/stressful experiences, feeding patterns, and other clinical and demographic data were collected. Gut microbiota was profiled using 16S rRNA sequencing, and operational taxonomic units (OTUs) were selected to predict the neurobehaviors. The neurobehavioral development was assessed by the Neonatal Neurobehavioral Scale (NNNS) at 36 to 38 weeks of post-menstrual age (PMA). Fifty-five infants who had NNNS measurements were included in the sparse log-contrast regression analysis. Results Preterm infants who experienced high level of pain/stress during the NICU hospitalization that were associated with higher NNNS stress/abstinence scores. Eight operational taxonomic units (OTUs) were identified to be associated with of NNNS subscales after controlling demographic and clinical features, feeding patterns, and painful/stressful experiences. These OTUs, taxa belong to seven genera including Enterobacteriaceae_unclassified, Escherichia-Shigella, Incertae_Sedis, Veillonella, Enterococcus, Clostridium_sensu_stricto_1 , and Streptococcus with five belonging to Firmicutes and two belonging to Proteobacteria phylum. The enriched abundance of Enterobacteriaceae_unclassified (OTU17) and Streptococcus (OTU28) were consistently associated with less optimal neurobehavioral outcomes. The other six OTUs were also associated with infant neurobehavioral responses depending on days at NICU stay. Conclusions This study explored the dynamic impact of specific OTUs on neurobehavioral development among preterm infants after controlling for early life experiences, i.e., acute and chronic pain/stress, and feeding in the NICU.
Collapse
Affiliation(s)
- Jie Chen
- Florida State University College of Nursing, Tallahassee, FL., United States
- School of Nursing, University of Connecticut, Storrs, CT., United States
| | - Hongfei Li
- Department of Statistics, University of Connecticut, Storrs, CT., United States
| | - Tingting Zhao
- School of Nursing, University of Connecticut, Storrs, CT., United States
| | - Kun Chen
- Department of Statistics, University of Connecticut, Storrs, CT., United States
| | - Ming-Hui Chen
- Department of Statistics, University of Connecticut, Storrs, CT., United States
| | - Zhe Sun
- Department of Statistics, University of Connecticut, Storrs, CT., United States
- Department of Biostatistics, Yale School of Public Health, New Haven, CT., United States
| | - Wanli Xu
- School of Nursing, University of Connecticut, Storrs, CT., United States
| | - Kendra Maas
- University of Connecticut, Microbial Analysis, Resources, and Services (MARS), Storrs, CT., United States
| | - Barry Lester
- Brown Center for the Study of Children at Risk, Departments of Psychiatry and Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI., United States
| | - Xiaomei Cong
- School of Nursing, University of Connecticut, Storrs, CT., United States
- Yale University School of Nursing, Orange, CT., United States
- Institute for Systems Genomics, University of Connecticut, Farmington, CT., United States
| |
Collapse
|
150
|
Xi M, Na X, Ma X, Lan H, Sun T, Liu WH, Hung W, Zhao A. Maternal diet associated with infants' intestinal microbiota mediated by predominant long-chain fatty acid in breast milk. Front Microbiol 2023; 13:1004175. [PMID: 36687649 PMCID: PMC9852834 DOI: 10.3389/fmicb.2022.1004175] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/09/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Long-chain fatty acids in breast milk are affected by the mother's diet and play an important role in the growth, development, and immune construction of infants. This study aims to explore the correlation between maternal diet, breast milk fatty acids (FAs), and the infant intestinal flora. Methods We enrolled 56 paired mothers and their infants; both breast milk samples and infants' fecal samples were collected to determine the long-chain FA content of breast milk by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS), and metagenomic technology was applied to determine the microbial composition of infant feces. The maternal diet was also investigated using a 24-h dietary recall. Results The results indicated that the fat contribution rates of edible oils in the maternal diet are significantly positively correlated with the contents of certain long-chain fatty acids (C16:0, C18:1, C16:1, and C22:4) in breast milk, which mainly regulate the abundance of Lacticaseibacillus rhamnosus, Lacticaseibacillus fermentum, and Lacticaseibacillus paracasei in the infant gut. Through KEGG pathway analysis, our data revealed that the long-chain FAs in different groups of breast milk were significantly correlated with the pathways of biotin metabolism, glycerolipid metabolism, and starch and sucrose metabolism. Discussion The results of this study suggest a pathway in which the diets of lactating mothers may affect the composition of the infant intestinal microbiota by influencing breast milk FAs and then further regulating infant health.
Collapse
Affiliation(s)
- Menglu Xi
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Xiaona Na
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Xia Ma
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China,Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Hanglian Lan
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China,Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Ting Sun
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China,Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Wei-Hsien Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China,Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Weilian Hung
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China,Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China,*Correspondence: Weilian Hung,
| | - Ai Zhao
- Vanke School of Public Health, Tsinghua University, Beijing, China,Ai Zhao,
| |
Collapse
|