151
|
Pivrncova E, Kotaskova I, Thon V. Neonatal Diet and Gut Microbiome Development After C-Section During the First Three Months After Birth: A Systematic Review. Front Nutr 2022; 9:941549. [PMID: 35967823 PMCID: PMC9364824 DOI: 10.3389/fnut.2022.941549] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Cesarean section (C-section) delivery imprints fundamentally on the gut microbiota composition with potential health consequences. With the increasing incidence of C-sections worldwide, there is a need for precise characterization of neonatal gut microbiota to understand how to restore microbial imbalance after C-section. After birth, gut microbiota development is shaped by various factors, especially the infant’s diet and antibiotic exposure. Concerning diet, current research has proposed that breastfeeding can restore the characteristic gut microbiome after C-section. Objectives In this systematic review, we provide a comprehensive summary of the current literature on the effect of breastfeeding on gut microbiota development after C-section delivery in the first 3 months of life. Methods The retrieved data from PubMed, Scopus, and Web of Science were evaluated according to the PICO/PECO strategy. Quality assessment was conducted by the Newcastle–Ottawa Scale. Results After critical selection, we identified 14 out of 4,628 studies for the evaluation of the impact of the diet after C-section delivery. The results demonstrate consistent evidence that C-section and affiliated intrapartum antibiotic exposure affect Bacteroidetes abundance and the incapacity of breastfeeding to reverse their reduction. Furthermore, exclusive breastfeeding shows a positive effect on Actinobacteria and Bifidobacteria restoration over the 3 months after birth. None of the included studies detected any significant changes in Lactobacillus abundance in breastfed infants after C-section. Conclusion C-section and intrapartum antibiotic exposure influence an infant’s gut microbiota by depletion of Bacteroides, regardless of the infant’s diet in the first 3 months of life. Even though breastfeeding increases the presence of Bifidobacteria, further research with proper feeding classification is needed to prove the restoration effect on some taxa in infants after C-section. Systematic Review Registration: [www.crd.york.ac.uk/prospero/], identifier [CRD42021287672].
Collapse
Affiliation(s)
- Eliska Pivrncova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Iva Kotaskova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vojtech Thon
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
152
|
Kapoor B, Gulati M, Rani P, Gupta R. Psoriasis: Interplay between dysbiosis and host immune system. Clin Exp Rheumatol 2022; 21:103169. [PMID: 35964945 DOI: 10.1016/j.autrev.2022.103169] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
With advancement in human microbiome research, an increasing number of scientific evidences have endorsed the key role of both gut and skin microbiota in the pathogenesis of psoriasis. Microbiome dysbiosis, characterized by altered diversity and composition, as well as rise of pathobionts, have been identified as possible triggers for recurrent episodes of psoriasis. Mechanistically, gut dysbiosis leads to "leaky gut syndrome" via disruption of epithelial bilayer, thereby, resulting in translocation of bacteria and other endotoxins to systemic circulation, which in turn, results in inflammatory response. Similarly, skin dysbiosis disrupts the cutaneous homeostasis, leading to invasion of bacteria and other pathogens to deeper layers of skin or even systemic circulation further enhanced by injury caused by pruritus-induced scratching, and elicit innate and adaptive inflammation. The present review explores the correlation of both skin and gut microbiota dysbiosis with psoriasis. Also, the studies highlighting the potential of bacteriotherapeutic approaches including probiotics, prebiotics, metabiotics, and fecal microbiota transplantation for the management of psoriasis have been discussed.
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.
| | - Pooja Rani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
153
|
Epidemiological Studies of Children's Gut Microbiota: Validation of Sample Collection and Storage Methods and Microbiota Analysis of Toddlers' Feces Collected from Diapers. Nutrients 2022; 14:nu14163315. [PMID: 36014821 PMCID: PMC9416069 DOI: 10.3390/nu14163315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The composition of human gut microbiota influences human health and disease over the long term. Since the flora in specimens can easily change at ambient temperature outside the body, epidemiological studies need feasible methods of stool specimen collection and storage to be established. We aimed to validate two methods: feces frozen-stored in tubes containing guanidine thiocyanate solution for two months after collection (Method B), and feces excreted in diapers and frozen-stored (Method C). Validation was by comparison with a gold standard Method A. Bacterial flora of five adults were sampled and stored by all three methods. Bacterial composition was examined by amplicon sequencing analysis. Bland-Altman analyses showed that Methods B and C might change relative abundances of certain bacterial flora. Thereafter, we analyzed the bacterial flora of 76 toddlers (two age groups) in stools sampled and processed by Method C. The diversity indices of toddlers' flora were less than those of adults. The relative abundance of some bacteria differed significantly between children aged 1.5 and 3 years. The specimen collection and storage methods validated in this study are worth adopting in large-scale epidemiological studies, especially for small children, provided the limited accuracy for some specific bacteria is understood.
Collapse
|
154
|
Zhang Z, Huang B, Wang Y, Zhu M, Wang C. Could Weaning Remodel the Oral Microbiota Composition in Donkeys? An Exploratory Study. Animals (Basel) 2022; 12:ani12162024. [PMID: 36009615 PMCID: PMC9404433 DOI: 10.3390/ani12162024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
As the initiation point of digestion, the oral microbiome is important in maintaining oral and systemic health. However, the composition of oral microbial communities and the influence of weaning on the oral microbiota of donkey foals remains poorly characterized. The present study used buccal swab samples to determine the changes in oral microbial communities occurring at the time of weaning. A total of 20 oral swab samples were collected from two groups: preweaning donkey foals (PreW group, n = 10) and postweaning donkey foals (PostW group, n = 10). The donkey oral microbiome was analyzed by 16S rRNA gene sequencing using Illumina MiSeq. This study is the first report of the donkey oral microbiome in association with weaning. Compared to the preweaning donkeys, the oral bacteria diversity in the postweaning donkeys was increased, with a higher Simpson index. Changes in the composition of the oral microbiota between the PreW and PostW groups were observed in the present study. At the phylum level, the relative abundance of Firmicutes and Myxococcota was significantly greater in the PostW than in the PreW group. At the genus level, the Gemella, unclassified_o__Lactobacillales, and Lactobacillus were increased in the postweaning donkeys. The donkeys’ oral microbial functions were predicted using PICRUSt, and the functions related to carbohydrate metabolic pathways were significantly enriched in the oral microbiome in the PostW donkeys. In summary, the current study provides a deeper insight into the oral microbiota changes during the weaning period, and the influence of weaning together with the documented changes in diversity and composition will help us to obtain a better understanding of their long-term health impact within the oral cavities of donkey foals. However, a major limitation of the present study was that the samples were obtained from different animals in the PreW and PostW groups, which may have resulted in variability among the different individuals. Further investigation is needed to monitor the shift in oral microbes in the same individuals during the weaning period.
Collapse
|
155
|
Li P, Chang X, Chen X, Tang T, Liu Y, Shang Y, Qi K. Dynamic colonization of gut microbiota and its influencing factors among the breast-feeding infants during the first two years of life. J Microbiol 2022; 60:780-794. [DOI: 10.1007/s12275-022-1641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
|
156
|
Currell A, Koplin JJ, Lowe AJ, Perrett KP, Ponsonby AL, Tang MLK, Dharmage SC, Peters RL. Mode of Birth Is Not Associated With Food Allergy Risk in Infants. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2135-2143.e3. [PMID: 35597762 DOI: 10.1016/j.jaip.2022.03.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The association between mode of delivery and the risk of food allergy remains unclear due to the absence of studies with both challenge-proven food allergy outcomes and detailed information on the type of caesarean delivery. OBJECTIVE We assessed whether emergency or elective caesarean, or caesarean delivery in the presence or absence of labor initiation, is associated with the risk of food allergy. METHODS The HealthNuts study recruited 5276 12-month-old infants who underwent skin prick testing and oral food challenge to ascertain food allergy status, and linked the child's study data to additional birth data from the Victorian Perinatal Data Collection. RESULTS Parents of 3006 children consented to data linkage, and birth data were obtained on 2045. In this subgroup, 30% were born by caesarean and 13% had food allergy. Caesarean delivery, compared with vaginal birth, was not associated with the risk of food allergy (adjusted odds ratio [aOR]: 0.95, 95% confidence interval [CI]: 0.70, 1.30). Neither caesarean delivery before the onset of labor, nor after the onset of labor, was associated with the risk of food allergy (aOR: 0.83, 95% CI: 0.55-1.23 and aOR: 1.13, 95% CI: 0.75-1.72, respectively). Delivery by elective or emergency caesarean, compared with vaginal delivery, was not associated with risk of food allergy (aOR: 1.05, 95% CI: 0.71-1.55 and aOR: 0.86, 95% CI: 0.56-1.31). There was no evidence of effect modification by breastfeeding, older siblings, pet dog ownership, or maternal allergy. CONCLUSION Caesarean delivery, either with or without labor, or elective or emergency, was not associated with the risk of food allergy in a population-based cohort of 12-month-old infants.
Collapse
Affiliation(s)
- Anne Currell
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Jennifer J Koplin
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Adrian J Lowe
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Kirsten P Perrett
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; Department of Allergy and Immunology, Royal Children's Hospital, Parkville, VIC, Australia
| | - Anne-Louise Ponsonby
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia; Neuroepidemiology Group, The Florey Institute for Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Mimi L K Tang
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia; Department of Allergy and Immunology, Royal Children's Hospital, Parkville, VIC, Australia
| | - Shyamali C Dharmage
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Rachel L Peters
- Population Health, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
157
|
The microbiome modulating potential of superheated steam (SHS) treatment of dietary fibres. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2022.103082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
158
|
Elliott MJ, Golombek SG. Evolution of Preterm Infant Nutrition from Breastfeeding to an Exclusive Human Milk Diet: A Review. Neoreviews 2022; 23:e558-e571. [PMID: 35909104 DOI: 10.1542/neo.23-8-e558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The benefits of feeding human milk to human infants are well-established. Preterm infants, particularly those born with very low birthweight (VLBW; <1,500 g), are a uniquely vulnerable population at risk for serious, life-threatening complications as well as disruptions in normal growth and development that can affect their lives into adulthood. Feeding VLBW preterm infants an exclusive human milk diet (EHMD) from birth that consists of the mother's own milk or donor human milk plus a nutritional fortifier made exclusively from human milk has been associated with a reduction in morbidity and mortality and improved early growth and developmental metrics. Preliminary evidence suggests that the health benefits of adopting an EHMD (or avoiding cow milk products) early in life may last into adulthood. This review briefly summarizes the history of breastfeeding and describes the available evidence on the benefits of an EHMD among VLBW preterm infants as well as the importance of high-quality manufacturing standards for producing safe and effective human milk-based products.
Collapse
Affiliation(s)
- Melinda J Elliott
- Department of Neonatology, Pediatrix Medical Group of Maryland, Rockville, MD
| | - Sergio G Golombek
- Prolacta Bioscience, Duarte, CA.,Departments of Neonatology and Pediatrics, SUNY Downstate Health Sciences University, Brooklyn, NY
| |
Collapse
|
159
|
Li J, Qu Z, Liu F, Jing H, Pan Y, Guo S, Ho CL. Diet‐Based Microbiome Modulation: You are What You Eat. PRINCIPLES IN MICROBIOME ENGINEERING 2022:1-46. [DOI: 10.1002/9783527825462.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
160
|
Wicaksono WA, Buko A, Kusstatscher P, Sinkkonen A, Laitinen OH, Virtanen SM, Hyöty H, Cernava T, Berg G. Modulation of the food microbiome by apple fruit processing. Food Microbiol 2022; 108:104103. [DOI: 10.1016/j.fm.2022.104103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/04/2022]
|
161
|
Reynoso-García J, Miranda-Santiago AE, Meléndez-Vázquez NM, Acosta-Pagán K, Sánchez-Rosado M, Díaz-Rivera J, Rosado-Quiñones AM, Acevedo-Márquez L, Cruz-Roldán L, Tosado-Rodríguez EL, Figueroa-Gispert MDM, Godoy-Vitorino F. A complete guide to human microbiomes: Body niches, transmission, development, dysbiosis, and restoration. FRONTIERS IN SYSTEMS BIOLOGY 2022; 2:951403. [PMID: 38993286 PMCID: PMC11238057 DOI: 10.3389/fsysb.2022.951403] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Humans are supra-organisms co-evolved with microbial communities (Prokaryotic and Eukaryotic), named the microbiome. These microbiomes supply essential ecosystem services that play critical roles in human health. A loss of indigenous microbes through modern lifestyles leads to microbial extinctions, associated with many diseases and epidemics. This narrative review conforms a complete guide to the human holobiont-comprising the host and all its symbiont populations- summarizes the latest and most significant research findings in human microbiome. It pretends to be a comprehensive resource in the field, describing all human body niches and their dominant microbial taxa while discussing common perturbations on microbial homeostasis, impacts of urbanization and restoration and humanitarian efforts to preserve good microbes from extinction.
Collapse
Affiliation(s)
| | | | | | - Kimil Acosta-Pagán
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | - Mitchell Sánchez-Rosado
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | - Jennifer Díaz-Rivera
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | - Angélica M. Rosado-Quiñones
- Department of Biology, UPR Rio Piedras Campus, San Juan, PR, United States
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | - Luis Acevedo-Márquez
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | - Lorna Cruz-Roldán
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| | | | | | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, UPR School of Medicine, San Juan, PR, United States
| |
Collapse
|
162
|
Kumari R, Yadav Y, Misra R, Das U, Das Adhikari U, Malakar P, Dubey GP. Emerging frontiers of antibiotics use and their impacts on the human gut microbiome. Microbiol Res 2022; 263:127127. [PMID: 35914416 DOI: 10.1016/j.micres.2022.127127] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/17/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023]
Abstract
Antibiotics, the primary drugs used to cure bacterial diseases, are increasingly becoming ineffective due to the emergence of multiple drug resistance (MDR) leading to recurrence of previously sensitive pathogens. Human gut microbiome (GM), known to play an important role in various physiological processes, consists of pool of diverse microbes. Indiscriminate use of antibiotics during the life span of an individual may lead to development of resistant microbes e.g. Vibrio, Acinetobacter, Escherichia, Klebsiella, Clostridia, etc. in the human GM. Transmission of antibiotic resistant genes (ARGs) between pathogenic and commensal bacteria occurs more frequently in microbiome communities wherein bacteria communicate and exchange cellular constituents both among themselves and with the host. Additionally, co-factors like 'early vs. late' exposure, type of antibiotics and duration of treatment modulate the adverse effects of antibiotics on GM maturation. Furthermore, factors like mode of birth, ethnicity, malnutrition, demography, diet, lifestyle, etc., which influence GM composition, can also indirectly alter the host response to antibiotics. Currently, advanced 'omics' and culturomics approaches are revealing novel avenues to study the interplay between antibiotics and the microbiome and to identify resistant genes in these bacterial communities. Here, we discuss the recent developments that have given insights into the effects of antibiotics on the homeostatic balance of the gut microbiome and thus on human health.
Collapse
Affiliation(s)
- Rekha Kumari
- Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Yasha Yadav
- Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Richa Misra
- Department of Zoology, Sri Venkateswara College, University of Delhi, Delhi 1100021, India
| | - Utpal Das
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Upasana Das Adhikari
- The Ragon Institute of MGH, MIT and Harvard, 400 Technology Square Cambridge, MA 02139, USA
| | - Pushkar Malakar
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gyanendra P Dubey
- Molecular Microbial Pathogenesis Unit, Institut Pasteur, 28 rue du Docteur Roux, 75724 Cedex 15 Paris, France.
| |
Collapse
|
163
|
The Ameliorating Effect of Lizhong-Tang on Antibiotic-Associated Imbalance in the Gut Microbiota in Mouse. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12146943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Some herbal medicines have anti-inflammatory and anti-diarrheal effects. This study analyzed the modulating effect of gut microbiota of anti-inflammatory herbal medicines on antibiotic-associated diarrhea (AAD). The anti-inflammatory effects of 10 herbal medicines and Lizhong-tang active compounds were studied by measuring the nitric oxide production in an in vitro experiment. This was followed by an in vivo experiment in which the anti-diarrheal effects of Lizhong-tang and Magnolia officinalis in a lincomycin-induced AAD mouse model were measured. Changes in the intestinal microflora were observed using terminal restriction fragment length polymorphism analysis. Both Lizhong-tang and M. officinalis were effective against AAD, with Lizhong-tang’s anti-diarrheal effects being particularly effective. In addition, the active compounds of Lizhong-tang, liquiritin and 6-gingerol, inhibited the expression of inducible nitric oxide synthase and cyclooxygenase-2, thus showing an anti-inflammatory effect. Gut microbiota analysis showed that Lizhong-tang could alter the composition of the gut microbiota and ameliorated imbalance in the gut microbiota in a lincomycin-induced AAD mouse model.
Collapse
|
164
|
El Mouzan M, Al-Hussaini AA, Al Sarkhy A, Assiri A, Alasmi M. Intestinal microbiota profile in healthy Saudi children: The bacterial domain. Saudi J Gastroenterol 2022; 28:312-317. [PMID: 35848701 PMCID: PMC9408733 DOI: 10.4103/sjg.sjg_585_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Knowledge of microbiota in health is essential for clinical research on the role of microbiota in disease. We aimed to characterize the intestinal microbiota in healthy Saudi children. Methods In this community-based study, stool samples were collected from a randomly selected sample of 20 healthy school children of Saudi origin. The samples were frozen at -80°C till analysis. Bacterial DNA was isolated and libraries were prepared using the Illumina Nextera XT library preparation kit. Unassembled sequencing reads were directly analyzed and quantified for each organism's relative abundance. The abundance for each organism was calculated and expressed as the average relative percentage from phyla to species. Results The median age was 11.3 (range 6.8-15.4) years, and 35% of them were males. The three most abundant phyla were Firmicutes, Bacteroidetes, and Actinobacteria accounting for 49%, 26%, and 24%, respectively. The most abundant genera included Bifidobacterium, Bacteroides, and Blautia accounting for 18.9%, 12.8%, and 8.2%, respectively. Finally, the most abundant species included 14 species belonging to the genus Bacteroides and nine species belonging to Bifidobacterium. Conclusions The abundance of intestinal microbiome in healthy Saudi children is different from that of other populations. Further studies are needed to understand the causes of variation between populations, which might lead to new preventive methods and treatment strategies of diseases caused by microbial dysbiosis.
Collapse
Affiliation(s)
- Mohammad El Mouzan
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulrahman A Al-Hussaini
- Division of Pediatric Gastroenterology, Children's Specialized Hospital, King Fahad Medical City; Faculty of Medicine, AlFaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Al Sarkhy
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Asaad Assiri
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mona Alasmi
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
165
|
Khanna HN, Roy S, Shaikh A, Bandi V. Emerging Role and Place of Probiotics in the Management of Pediatric Neurodevelopmental Disorders. Euroasian J Hepatogastroenterol 2022; 12:102-108. [PMID: 36959989 PMCID: PMC10028704 DOI: 10.5005/jp-journals-10018-1384] [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: 03/16/2023] Open
Abstract
The current decade has witnessed significant developments with the latest therapeutic agents for managing various infectious diseases to complex hemato-oncological conditions, leading to a decrease in morbidity and mortality, while improving the quality of life (QoL), and increasing the life span. Non-communicable diseases (NCDs), which are on the rise across all age-groups, are being driven by unhealthy lifestyles and improved mental health issues. The current therapeutic agents were found to offer only symptomatic relief of varying efficacy and significant adverse effects, leading clinicians to evaluate other options for the management of both neurodevelopmental and neurodegenerative disorders. The role of gut microbiota has emerged as a potential target for the treatment of both neurodegenerative diseases and neurodevelopmental disorders like attention-deficit hyperactivity disorder (ADHD)/autism spectrum disorders (ASD) as a result of the decoding of the human genome and advances in our understanding of the human gut microbiome, including its interactions with the human brain. This review has been undertaken to understand on date level of understanding of human microbiota and towards identifying probiotic strains with proven efficacy and safety. According to recent investigations, several lactobacillus strains, including L. Paracasei 37, L. Planetarium 128, L. reuteri DSM 17938, and Bifidobacterium longum, have been effective in treating children's neurodevelopmental disorders such as ASD and ADHD. Future clinical studies are nonetheless required to confirm the long-term safety and effectiveness of probiotic strains in managing the primary and comorbid symptoms, hence improving patient and family quality of life. How to cite this article Khanna HN, Roy S, Shaikh A, et al. Emerging Role and Place of Probiotics in the Management of Pediatric Neurodevelopmental Disorders. Euroasian J Hepato-Gastroenterol 2022;12(2):102-108.
Collapse
Affiliation(s)
- Himani Narula Khanna
- Department of community Medicine, HIMSR, Jamia-Hamdard University, New Delhi, India
| | - Sushovan Roy
- Department of Community Medicine, HIMSR, New Delhi, India
| | - Aqsa Shaikh
- Department of Community Medicine, HIMSR, New Delhi, India
| | - Viswanath Bandi
- Research Scholar, Faculty of management studies, ICFAI University, Ranchi, Jharkhand, India
| |
Collapse
|
166
|
Li X, Ren Y, Zhang J, Ouyang C, Wang C, Lu F, Yin Y. Development of Early-Life Gastrointestinal Microbiota in the Presence of Antibiotics Alters the Severity of Acute DSS-Induced Colitis in Mice. Microbiol Spectr 2022; 10:e0269221. [PMID: 35438531 PMCID: PMC9241904 DOI: 10.1128/spectrum.02692-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/30/2022] [Indexed: 12/05/2022] Open
Abstract
Early-life gastrointestinal microbiota development is crucial for physiological development and immunological homeostasis. In the current study, perinatal microbiota and the development of gastrointestinal microbiota in different early-life periods (perinatal, lactation, and postweaning nutrition periods) were explored by using an antibiotic-interfered mouse model and a dextran sulfate sodium-induced colitis mouse model. Gut microbiota samples were collected from mother mice and litters. The results of 16S rRNA gene sequences suggested that microbiota in the gastrointestinal system were present in prenatal fetal mice, and microbiota structures in different parts of the gastrointestinal system of the fetal mice were similar to those in the corresponding gut parts of maternal mice. Microbiota in mucus samples from different regions exhibited higher diversity at birth than at other periods and varied substantially over time with diet change. Moreover, antibiotic treatment in early life affected the composition and diversity of gastrointestinal microbiota in adult mice and enhanced susceptibility to experimental colitis in mice, particularly in the lactation period. This approach of exploring gut microbiota evolution is hoped to provide an enhanced view of how resident microbiota develop in early life, which in turn might facilitate understanding of gut microbiota and related diseases. IMPORTANCE This study investigated resident microbiota in the whole gastrointestinal (GI) tract to explore gut microbiota development in early life and found that early-life antibiotic exposure exacerbated alterations in gut microbiota and murine dextran sulfate sodium (DSS)-induced colitis. Furthermore, the presence of bacteria in the GI tract of mice before birth and the importance of the lactation period in GI microbiota development were confirmed.
Collapse
Affiliation(s)
- Xiaojun Li
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yu Ren
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Jie Zhang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chunhui Ouyang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chunlian Wang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Fanggen Lu
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, and Research Center of Digestive Disease, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yani Yin
- Department of Gastroenterology, Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, People’s Republic of China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
167
|
Bendová B, Mikula O, Bímová BV, Čížková D, Daniszová K, Ďureje Ľ, Hiadlovská Z, Macholán M, Martin JF, Piálek J, Schmiedová L, Kreisinger J. Divergent gut microbiota in two closely related house mouse subspecies under common garden conditions. FEMS Microbiol Ecol 2022; 98:6620832. [PMID: 35767862 DOI: 10.1093/femsec/fiac078] [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: 12/03/2021] [Revised: 06/02/2022] [Accepted: 06/27/2022] [Indexed: 11/14/2022] Open
Abstract
The gastrointestinal microbiota (GM) is considered an important component of the vertebrate holobiont. GM-host interactions influence the fitness of holobionts and are therefore an integral part of evolution. The house mouse is a prominent model for GM-host interactions, and evidence suggests a role for GM in mouse speciation. However, previous studies based on short 16S rRNA GM profiles of wild house mouse subspecies failed to detect GM divergence, which is a prerequisite for the inclusion of GM in Dobzhansky-Muller incompatibilities. Here, we used standard 16S rRNA GM profiling in two mouse subspecies, Mus musculus musculus and M. m. domesticus, including the intestinal mucosa and content of three gut sections (ileum, caecum, and colon). We reduced environmental variability by sampling GM in the offspring of wild mice bred under semi-natural conditions. Although the breeding conditions allowed a contact between the subspecies, we found a clear differentiation of GM between them, in all three gut sections. Differentiation was mainly driven by several Helicobacters and two H. ganmani variants showed a signal of co-divergence with their hosts. Helicobacters represent promising candidates for studying GM-host co-adaptations and the fitness effects of their interactions.
Collapse
Affiliation(s)
- Barbora Bendová
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic.,Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Ondřej Mikula
- Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic.,Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | | | - Dagmar Čížková
- Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Kristina Daniszová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Ľudovít Ďureje
- Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Zuzana Hiadlovská
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Miloš Macholán
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | | | - Jaroslav Piálek
- Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Lucie Schmiedová
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic.,Studenec Research Facility, Institute of Vertebrate Biology, Czech Academy of Sciences, Brno, Czech Republic
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
168
|
Kato S, Nagasawa T, Uehara O, Shimizu S, Sugiyama N, Hasegawa-Nakamura K, Noguchi K, Hatae M, Kakinoki H, Furuichi Y. Increase in Bifidobacterium is a characteristic of the difference in the salivary microbiota of pregnant and non-pregnant women. BMC Oral Health 2022; 22:260. [PMID: 35764953 PMCID: PMC9238123 DOI: 10.1186/s12903-022-02293-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/21/2022] [Indexed: 12/30/2022] Open
Abstract
Background The establishment of symbiotic microbiota in pregnant women is important for both the mother and her offspring. Little is known about the salivary symbiotic bacteria in pregnancy, and analysis of composition of microbiome (ANCOM) is useful to detect small differences in the number of bacteria. The aim of this study was to investigate the differences in the salivary bacteria between healthy pregnant and non-pregnant women using ANCOM. Methods Unstimulated saliva samples were collected from 35 healthy pregnant women at 35 weeks gestation and 30 healthy non-pregnant women during menstruation. All participants underwent a periodontal examination. Estradiol and progesterone levels were examined by enzyme-linked immunosorbent assay. DNA extracted from the saliva was assessed by 16S ribosomal RNA amplicon sequencing and real-time PCR. Results Salivary estradiol and progesterone levels were significantly increased in pregnant women. The alpha and beta diversities were higher in pregnant women than in non-pregnant women. The largest effect size difference noted when the microbiota of the pregnant and non-pregnant women were analyzed was that for Bifidobacteriales. Levels of Bifidobacterium dentium, but not of Bifidobacterium adolescentis, were significantly increased in pregnant women, and the levels were significantly correlated with progesterone concentration. Conclusion The results suggest that Bifidobacterium and progesterone levels are elevated in the saliva of healthy pregnant women compared with non-pregnant women.
Collapse
|
169
|
Boudar Z, Sehli S, El Janahi S, Al Idrissi N, Hamdi S, Dini N, Brim H, Amzazi S, Nejjari C, Lloyd-Puryear M, Ghazal H. Metagenomics Approaches to Investigate the Neonatal Gut Microbiome. Front Pediatr 2022; 10:886627. [PMID: 35799697 PMCID: PMC9253679 DOI: 10.3389/fped.2022.886627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
Early infancy is critical for the development of an infant's gut flora. Many factors can influence microbiota development during the pre- and postnatal periods, including maternal factors, antibiotic exposure, mode of delivery, dietary patterns, and feeding type. Therefore, investigating the connection between these variables and host and microbiome interactions in neonatal development would be of great interest. As the "unculturable" era of microbiome research gives way to an intrinsically multidisciplinary field, microbiome research has reaped the advantages of technological advancements in next-generation sequencing, particularly 16S rRNA gene amplicon and shotgun sequencing, which have considerably expanded our knowledge about gut microbiota development during early life. Using omics approaches to explore the neonatal microbiome may help to better understand the link between the microbiome and newborn diseases. Herein, we summarized the metagenomics methods and tools used to advance knowledge on the neonatal microbiome origin and evolution and how the microbiome shapes early and late individuals' lives for health and disease. The way to overcome limitations in neonatal microbiome studies will be discussed.
Collapse
Affiliation(s)
- Zakia Boudar
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sofia Sehli
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Sara El Janahi
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Najib Al Idrissi
- Department of Surgery, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Salsabil Hamdi
- Laboratory of Genomics and Bioinformatics, School of Pharmacy, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Nouzha Dini
- Mother and Child Department, Cheikh Khalifa International University Hospital, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Hassan Brim
- Department of Pathology, Howard University, Washington, DC, United States
| | - Saaïd Amzazi
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Chakib Nejjari
- Department of Epidemiology and Biostatistics, International School of Public Health, Mohammed VI University of Health Sciences, Casablanca, Morocco
- Department of Epidemiology and Public Health, Faculty of Medicine, University Sidi Mohammed Ben Abdellah, Fez, Morocco
| | | | - Hassan Ghazal
- Department of Fundamental Sciences, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
- National Center for Scientific and Technical Research, Rabat, Morocco
| |
Collapse
|
170
|
Selvakumar D, Evans D, Coyte KZ, McLaughlin J, Brass A, Hancock L, Cruickshank S. Understanding the development and function of the gut microbiota in health and inflammation. Frontline Gastroenterol 2022; 13:e13-e21. [PMID: 35812026 PMCID: PMC9234741 DOI: 10.1136/flgastro-2022-102119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
The gut microbiota is known to play an important role in maintaining gut health through a symbiotic relationship with the host. Altered gut microbiota is a common feature of several diseases of the gastrointestinal tract; however, the causal relationship between microbiota and disease pathogenesis is poorly understood. Necrotising enterocolitis (NEC) and inflammatory bowel disease (IBD) are both severe inflammatory diseases affecting the gastrointestinal tract. Although they affect very different patient populations, with NEC primarily being a disease of prematurity and IBD predominantly affecting adults although children can be affected, they both demonstrate common features of gut microbial dysbiosis and a dysregulated host immune response. By comparing and contrasting the changes in gut microbiota, host immune response and function, we aim to highlight common features in diseases that may seem clinically unrelated. Key areas of interest are the role of pattern recognition receptors in altered recognition and responses to the gut microbiota by the host immune system and the associated dysfunctional gut epithelial barrier. The challenge of identifying causal relationships between microbiota and disease is ever-present; however, considering a disease-agnostic approach may help to identify mechanistic pathways shared across several clinical diseases.
Collapse
Affiliation(s)
- Deepak Selvakumar
- Department of Colorectal Surgery, Manchester University NHS Foundation Trust, Manchester, UK,Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Dolan Evans
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Katharine Z Coyte
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - John McLaughlin
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK,Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Andy Brass
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Laura Hancock
- Department of Colorectal Surgery, Manchester University NHS Foundation Trust, Manchester, UK,Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sheena Cruickshank
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
171
|
Angkeow JW, Monaco DR, Chen A, Venkataraman T, Jayaraman S, Valencia C, Sie BM, Liechti T, Farhadi PN, Funez-dePagnier G, Sherman-Baust CA, Wong MQ, Ruczinski I, Caturegli P, Sears CL, Simner PJ, Round JL, Duggal P, Laserson U, Steiner TS, Sen R, Lloyd TE, Roederer M, Mammen AL, Longman RS, Rider LG, Larman HB. Phage display of environmental protein toxins and virulence factors reveals the prevalence, persistence, and genetics of antibody responses. Immunity 2022; 55:1051-1066.e4. [PMID: 35649416 PMCID: PMC9203978 DOI: 10.1016/j.immuni.2022.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/17/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022]
Abstract
Microbial exposures are crucial environmental factors that impact healthspan by sculpting the immune system and microbiota. Antibody profiling via Phage ImmunoPrecipitation Sequencing (PhIP-Seq) provides a high-throughput, cost-effective approach for detecting exposure and response to microbial protein products. We designed and constructed a library of 95,601 56-amino acid peptide tiles spanning 14,430 proteins with "toxin" or "virulence factor" keyword annotations. We used PhIP-Seq to profile the antibodies of ∼1,000 individuals against this "ToxScan" library. In addition to enumerating immunodominant antibody epitopes, we studied the age-dependent stability of the ToxScan profile and used a genome-wide association study to find that the MHC-II locus modulates bacterial epitope selection. We detected previously described anti-flagellin antibody responses in a Crohn's disease cohort and identified an association between anti-flagellin antibodies and juvenile dermatomyositis. PhIP-Seq with the ToxScan library is thus an effective tool for studying the environmental determinants of health and disease at cohort scale.
Collapse
Affiliation(s)
- Julia W Angkeow
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Monaco
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Athena Chen
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Thiagarajan Venkataraman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sahana Jayaraman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cristian Valencia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brandon M Sie
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas Liechti
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Payam N Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH, Bethesda, MD, USA
| | - Gabriela Funez-dePagnier
- Jill Roberts Institute for Research in IBD, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cheryl A Sherman-Baust
- Laboratory of Molecular Biology and Immunology, NIH/National Institute on Aging, Baltimore, MD, USA
| | - May Q Wong
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Patrizio Caturegli
- Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Departments of Medicine and Oncology, Johns Hopkins University School of Medicine, and Department of Molecular Microbiology & Immunology, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Patricia J Simner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Uri Laserson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ranjan Sen
- Laboratory of Molecular Biology and Immunology, NIH/National Institute on Aging, Baltimore, MD, USA
| | - Thomas E Lloyd
- Department of Neurology, Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulations, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Randy S Longman
- Jill Roberts Institute for Research in IBD, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, NIH, Bethesda, MD, USA
| | - H Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
172
|
Wells JM, Gao Y, de Groot N, Vonk MM, Ulfman L, van Neerven RJJ. Babies, Bugs, and Barriers: Dietary Modulation of Intestinal Barrier Function in Early Life. Annu Rev Nutr 2022; 42:165-200. [PMID: 35697048 DOI: 10.1146/annurev-nutr-122221-103916] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The intestinal barrier is essential in early life to prevent infection, inflammation, and food allergies. It consists of microbiota, a mucus layer, an epithelial layer, and the immune system. Microbial metabolites, the mucus, antimicrobial peptides, and secretory immunoglobulin A (sIgA) protect the intestinal mucosa against infection. The complex interplay between these functionalities of the intestinal barrier is crucial in early life by supporting homeostasis, development of the intestinal immune system, and long-term gut health. Exclusive breastfeeding is highly recommended during the first 6 months. When breastfeeding is not possible, milk-based infant formulas are the only safe alternative. Breast milk contains many bioactive components that help to establish the intestinal microbiota and influence the development of the intestinal epithelium and the immune system. Importantly, breastfeeding lowers the risk for intestinal and respiratory tract infections. Here we review all aspects of intestinal barrier function and the nutritional components that impact its functionality in early life, such as micronutrients, bioactive milk proteins, milk lipids, and human milk oligosaccharides. These components are present in breast milk and can be added to milk-based infant formulas to support gut health and immunity. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jerry M Wells
- Host Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Yifan Gao
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands
| | | | | | | | - R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands.,FrieslandCampina, Amersfoort, The Netherlands;
| |
Collapse
|
173
|
Tuniyazi M, Li S, Hu X, Fu Y, Zhang N. The Role of Early Life Microbiota Composition in the Development of Allergic Diseases. Microorganisms 2022; 10:1190. [PMID: 35744708 PMCID: PMC9227185 DOI: 10.3390/microorganisms10061190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022] Open
Abstract
Allergic diseases are becoming a major healthcare issue in many developed nations, where living environment and lifestyle are most predominantly distinct. Such differences include urbanized, industrialized living environments, overused hygiene products, antibiotics, stationary lifestyle, and fast-food-based diets, which tend to reduce microbial diversity and lead to impaired immune protection, which further increase the development of allergic diseases. At the same time, studies have also shown that modulating a microbiocidal community can ameliorate allergic symptoms. Therefore, in this paper, we aimed to review recent findings on the potential role of human microbiota in the gastrointestinal tract, surface of skin, and respiratory tract in the development of allergic diseases. Furthermore, we addressed a potential therapeutic or even preventive strategy for such allergic diseases by modulating human microbial composition.
Collapse
Affiliation(s)
| | | | | | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.T.); (S.L.); (X.H.)
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130062, China; (M.T.); (S.L.); (X.H.)
| |
Collapse
|
174
|
Effects of Concentrate Supplementation on Growth Performance, Rumen Fermentation, and Bacterial Community Composition in Grazing Yaks during the Warm Season. Animals (Basel) 2022; 12:ani12111398. [PMID: 35681862 PMCID: PMC9179552 DOI: 10.3390/ani12111398] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 11/17/2022] Open
Abstract
This study aimed to evaluate the effects of concentrate supplementation on the growth performance, serum biochemical parameters, rumen fermentation, and bacterial community composition of grazing yaks during the warm season. Eight male yaks (body weight, 123.96 ± 7.43 kg; 3-years) were randomly allocated to two treatments groups: grazing (n = 4, GY) and concentrate supplement group (n = 4, GYS). Concentrate supplementation increased the average daily gain (ADG) (p < 0.05). Glucose (GLU), total protein (TP), and aspartate aminotransferase (AST) serum concentrations were significantly higher in the GYS group than in the GY group (p < 0.05). Ammonia-N, MCP: microbial protein, and total volatile fatty acid concentrations were significantly higher in the GYS group than in the GY group (p < 0.01), whereas the pH and acetate: propionate values were significantly decreased (p < 0.01). The relative abundance of Firmicutes in the rumen fluid was significantly higher in the GYS group than in the GY group (p < 0.01). At the genus level, the relative abundances of Succiniclasticum, Prevotellaceae_UCG_003, Prevotellaceae_UCG_005, and Ruminococcus_1 were significantly greater in the GY group than in the GYS group (p < 0.01). In conclusion, concentrate supplementation improved yaks’ growth potential during the warm season, improved ruminal fermentation, and altered core bacteria abundance.
Collapse
|
175
|
Fovet CM, Pimienta C, Galhaut M, Relouzat F, Nunez N, Cavarelli M, Sconosciuti Q, Dhooge N, Marzinotto I, Lampasona V, Tolazzi M, Scarlatti G, Ho Tsong Fang R, Naninck T, Dereuddre-Bosquet N, Van Wassenhove J, Gallouët AS, Maisonnasse P, Le Grand R, Menu E, Seddiki N. A Case Study to Dissect Immunity to SARS-CoV-2 in a Neonate Nonhuman Primate Model. Front Immunol 2022; 13:855230. [PMID: 35603150 PMCID: PMC9114777 DOI: 10.3389/fimmu.2022.855230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Most children are less severely affected by coronavirus-induced disease 2019 (COVID-19) than adults, and thus more difficult to study progressively. Here, we provide a neonatal nonhuman primate (NHP) deep analysis of early immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in blood and mucosal tissues. In addition, we provide a comparison with SARS-CoV-2-infected adult NHP. Infection of the neonate resulted in a mild disease compared with adult NHPs that develop, in most cases, moderate lung lesions. In concomitance with the viral RNA load increase, we observed the development of an early innate response in the blood, as demonstrated by RNA sequencing, flow cytometry, and cytokine longitudinal data analyses. This response included the presence of an antiviral type-I IFN gene signature, a persistent and lasting NKT cell population, a balanced peripheral and mucosal IFN-γ/IL-10 cytokine response, and an increase in B cells that was accompanied with anti-SARS-CoV-2 antibody response. Viral kinetics and immune responses coincided with changes in the microbiota profile composition in the pharyngeal and rectal mucosae. In the mother, viral RNA loads were close to the quantification limit, despite the very close contact with SARS-CoV-2-exposed neonate. This pilot study demonstrates that neonatal NHPs are a relevant model for pediatric SARS-CoV-2 infection, permitting insights into the early steps of anti-SARS-CoV-2 immune responses in infants.
Collapse
Affiliation(s)
- Claire-Maëlle Fovet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Camille Pimienta
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Mathilde Galhaut
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Francis Relouzat
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | | | - Mariangela Cavarelli
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Quentin Sconosciuti
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Nina Dhooge
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Monica Tolazzi
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Raphaël Ho Tsong Fang
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Jérôme Van Wassenhove
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France.,MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Nabila Seddiki
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| |
Collapse
|
176
|
Hu M, Alashkar Alhamwe B, Santner-Nanan B, Miethe S, Harb H, Renz H, Potaczek DP, Nanan RK. Short-Chain Fatty Acids Augment Differentiation and Function of Human Induced Regulatory T Cells. Int J Mol Sci 2022; 23:ijms23105740. [PMID: 35628549 PMCID: PMC9143307 DOI: 10.3390/ijms23105740] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Regulatory T cells (Tregs) control immune system activity and inhibit inflammation. While, in mice, short-chain fatty acids (SCFAs) are known to be essential regulators of naturally occurring and in vitro induced Tregs (iTregs), data on their contribution to the development of human iTregs are sparse, with no reports of the successful SCFAs-augmented in vitro generation of fully functional human iTregs. Likewise, markers undoubtedly defining human iTregs are missing. Here, we aimed to generate fully functional human iTregs in vitro using protocols involving SCFAs and to characterize the underlying mechanism. Our target was to identify the potential phenotypic markers best characterizing human iTregs. Naïve non-Treg CD4+ cells were isolated from the peripheral blood of 13 healthy adults and cord blood of 12 healthy term newborns. Cells were subjected to differentiation toward iTregs using a transforming growth factor β (TGF-β)-based protocol, with or without SCFAs (acetate, butyrate, or propionate). Thereafter, they were subjected to flow cytometric phenotyping or a suppression assay. During differentiation, cells were collected for chromatin-immunoprecipitation (ChIP)-based analysis of histone acetylation. The enrichment of the TGF-β-based protocol with butyrate or propionate potentiated the in vitro differentiation of human naïve CD4+ non-Tregs towards iTregs and augmented the suppressive capacity of the latter. These seemed to be at least partly underlain by the effects of SCFAs on the histone acetylation levels in differentiating cells. GITR, ICOS, CD39, PD-1, and PD-L1 were proven to be potential markers of human iTregs. Our results might boost the further development of Treg-based therapies against autoimmune, allergic and other chronic inflammatory disorders.
Collapse
Affiliation(s)
- Mingjing Hu
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Discipline of Obstetrics, Gynaecology and Neonatology, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia
- Nepean Hospital, Derby Street, Kingswood, NSW 2747, Australia
| | - Bilal Alashkar Alhamwe
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute for Tumor Immunology, Clinic for Hematology, Immunology, and Oncology, Philipps University Marburg, 35043 Marburg, Germany
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Brigitte Santner-Nanan
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
| | - Sarah Miethe
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany
| | - Hani Harb
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Institute of Medical Microbiology and Virology, Medical Faculty, Technische Universität Dresden, 01307 Dresden, Germany
| | - Harald Renz
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
| | - Daniel P. Potaczek
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Institute of Laboratory Medicine, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL), and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany;
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Philipps University of Marburg-Medical Faculty, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany
| | - Ralph K. Nanan
- Charles Perkins Centre Nepean, Sydney Medical School Nepean, The University of Sydney, Sydney, NSW 2747, Australia; (M.H.); (B.S.-N.)
- International Inflammation (in-VIVO) Network, Worldwide Universities Network (WUN), West New York, NJ 10001, USA; (B.A.A.); (H.H.); (H.R.); (D.P.P.)
- Correspondence: ; Tel.: +61-2-4734-2612; Fax: +61-2-4734-1144
| |
Collapse
|
177
|
Foster-Nyarko E, Pallen MJ. The microbial ecology of Escherichia coli in the vertebrate gut. FEMS Microbiol Rev 2022; 46:fuac008. [PMID: 35134909 PMCID: PMC9075585 DOI: 10.1093/femsre/fuac008] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli has a rich history as biology's 'rock star', driving advances across many fields. In the wild, E. coli resides innocuously in the gut of humans and animals but is also a versatile pathogen commonly associated with intestinal and extraintestinal infections and antimicrobial resistance-including large foodborne outbreaks such as the one that swept across Europe in 2011, killing 54 individuals and causing approximately 4000 infections and 900 cases of haemolytic uraemic syndrome. Given that most E. coli are harmless gut colonizers, an important ecological question plaguing microbiologists is what makes E. coli an occasionally devastating pathogen? To address this question requires an enhanced understanding of the ecology of the organism as a commensal. Here, we review how our knowledge of the ecology and within-host diversity of this organism in the vertebrate gut has progressed in the 137 years since E. coli was first described. We also review current approaches to the study of within-host bacterial diversity. In closing, we discuss some of the outstanding questions yet to be addressed and prospects for future research.
Collapse
Affiliation(s)
- Ebenezer Foster-Nyarko
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Mark J Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, United Kingdom
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, United Kingdom
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TU, United Kingdom
| |
Collapse
|
178
|
Gonçalves JIB, Borges TJ, de Souza APD. Microbiota and the Response to Vaccines Against Respiratory Virus. Front Immunol 2022; 13:889945. [PMID: 35603203 PMCID: PMC9122122 DOI: 10.3389/fimmu.2022.889945] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
This mini review describes the role of gut and lung microbiota during respiratory viral infection and discusses the implication of the microbiota composition on the immune responses generated by the vaccines designed to protect against these pathogens. This is a growing field and recent evidence supports that the composition and function of the microbiota can modulate the immune response of vaccination against respiratory viruses such as influenza and SARS-CoV-2. Recent studies have highlighted that molecules derived from the microbiome can have systemic effects, acting in distant organs. These molecules are recognized by the immune cells from the host and can trigger or modulate different responses, interfering with vaccination protection. Modulating the microbiota composition has been suggested as an approach to achieving more efficient protective immune responses. Studies in humans have reported associations between a better vaccine response and specific bacterial taxa. These associations vary among different vaccine strategies and are likely to be context-dependent. The use of prebiotics and probiotics in conjunction with vaccination demonstrated that bacterial components could act as adjuvants. Future microbiota-based interventions may potentially improve and optimize the responses of respiratory virus vaccines.
Collapse
Affiliation(s)
- João I. B. Gonçalves
- Laboratory of Clinical and Experimental Immunology, Health and Life Science School - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Thiago J. Borges
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Health and Life Science School - Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
179
|
Piawah S, Walker EJ, Van Blarigan EL, Atreya CE. The Gut Microbiome in Colorectal Cancer. Hematol Oncol Clin North Am 2022; 36:491-506. [DOI: 10.1016/j.hoc.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
180
|
Hanachi M, Maghrebi O, Bichiou H, Trabelsi F, Bouyahia NM, Zhioua F, Belghith M, Harigua-Souiai E, Baouendi M, Guizani-Tabbane L, Benkahla A, Souiai O. Longitudinal and Comparative Analysis of Gut Microbiota of Tunisian Newborns According to Delivery Mode. Front Microbiol 2022; 13:780568. [PMID: 35547149 PMCID: PMC9083410 DOI: 10.3389/fmicb.2022.780568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/15/2022] [Indexed: 12/01/2022] Open
Abstract
Microbiota colonization is a dynamic process that impacts the health status during an individual's lifetime. The composition of the gut microbiota of newborns is conditioned by multiple factors, including the delivery mode (DM). Nonetheless, the DM's influence remains uncertain and is still the subject of debate. In this context, the medical indication and the emergency of a cesarean delivery might have led to confounding conclusions regarding the composition and diversity of the neonatal microbiome. Herein, we used high-resolution shotgun sequencing to decipher the composition and dynamics of the gut microbiota composition of Tunisian newborns. Stool samples were collected from 5 elective cesarean section (ECS) and 5 vaginally delivered (VD) newborns at the following time points: Day 0, Day 15, and Day 30. The ECS and VD newborns showed the same level of bacterial richness and diversity. In addition, our data pointed to a shift in microbiota community composition during the first 2 weeks, regardless of the DM. Both ECS and VD showed a profile dominated by Proteobacteria, Actinobacteria, and Firmicutes. However, ECS showed an underrepresentation of Bacteroides and an enrichment of opportunistic pathogenic species of the ESKAPE group, starting from the second week. Besides revealing the intestinal microbiota of Tunisian newborns, this study provides novel insights into the microbiota perturbations caused by ECS.
Collapse
Affiliation(s)
- Mariem Hanachi
- Laboratory of Bioinformatics, bioMathematics and Biostatistics—LR16IPT09, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
- Faculty of Science of Bizerte, University of Carthage, Tunis, Tunisia
| | - Olfa Maghrebi
- Laboratory of Transmission, Control, and Immunobiology of Infections—LR16 IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology, and Biomolecules—LR16 IPT06, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Ferdaous Trabelsi
- Service de Gynécologie et Obstétrique, Hôpital Régional de Zaghouan, Zaghouan, Tunisia
| | - Najla Maha Bouyahia
- Service de Gynécologie Obstétrique et Médecine de la Reproduction, Hôpital Aziza Othmana, Tunis, Tunisia
| | - Fethi Zhioua
- Service de Gynécologie Obstétrique et Médecine de la Reproduction, Hôpital Aziza Othmana, Tunis, Tunisia
| | - Meriam Belghith
- Laboratory of Transmission, Control, and Immunobiology of Infections—LR16 IPT02, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Emna Harigua-Souiai
- Laboratory of Molecular Epidemiology and Experimental Pathology—LR16IPT04, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Meriem Baouendi
- Laboratory of Bioinformatics, bioMathematics and Biostatistics—LR16IPT09, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology, and Biomolecules—LR16 IPT06, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Alia Benkahla
- Laboratory of Bioinformatics, bioMathematics and Biostatistics—LR16IPT09, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Oussema Souiai
- Laboratory of Bioinformatics, bioMathematics and Biostatistics—LR16IPT09, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
181
|
Changes to Gut Microbiota Following Systemic Antibiotic Administration in Infants. Antibiotics (Basel) 2022; 11:antibiotics11040470. [PMID: 35453221 PMCID: PMC9025670 DOI: 10.3390/antibiotics11040470] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Long-term antibiotic use can have consequences on systemic diseases, such as obesity, allergy, and depression, implicating the causal role of gut microbiome imbalance. However, the evaluation of the effect of antibiotics in early infancy on alterations to the gut microbiome remains poorly understood. This study aimed to evaluate the gut microbiome state in infancy following systemic antibiotic treatment. Twenty infants under 3 months of age who had received antibiotics for at least 3 days were enrolled, and their fecal samples were collected 4 weeks after antibiotic administration finished. Thirty-four age-matched healthy controls without prior exposure to antibiotics were also assessed. The relative bacterial abundance in feces was obtained via sequencing of 16 S rRNA genes, and alpha and beta diversities were evaluated. At the genus level, the relative abundance of Escherichia/Shigella and Bifidobacterium increased (p = 0.03 and p = 0.017, respectively) but that of Bacteroides decreased (p = 0.02) in the antibiotic treatment group. The microbiome of the antibiotic treatment group exhibited an alpha diversity lower than that of the control group. Thus, systemic antibiotic administration in early infancy affects the gut microbiome composition even after a month has passed; long-term studies are needed to further evaluate this.
Collapse
|
182
|
Arepally D, Reddy RS, Goswami TK, Coorey R. A Review on Probiotic Microencapsulation and Recent Advances of their Application in Bakery Products. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-022-02796-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
183
|
Huang J, Zhang J, Wang X, Jin Z, Zhang P, Su H, Sun X. Effect of Probiotics on Respiratory Tract Allergic Disease and Gut Microbiota. Front Nutr 2022; 9:821900. [PMID: 35295917 PMCID: PMC8920559 DOI: 10.3389/fnut.2022.821900] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Allergy is a hypersensitivity reaction triggered by specific cell or antibody-mediated immune mechanisms. Allergies have increased in industrialized countries in recent decades. The rise in allergic respiratory diseases such as allergic rhinitis (AR) and allergic asthma (AA) is a potential threat to public health. Searches were conducted using PubMed, Google Scholar and Medline using the following key terms: allergic rhinitis OR asthma AND probiotics, allergic airway inflammation AND immune disorders, probiotics OR gut microbiota AND allergic disease, probiotics AND inflammatory. Studies from all years were included, specifically those published within the last 10 years. Some review articles and their reference lists were searched to identify related articles. The role of microbiota in respiratory allergic diseases has attracted more and more attention. Pieces of evidence suggested that the development of allergic diseases causes a possible imbalance in the composition of the gut microbiota. Compared to colonized mice, germ-free mice exhibit exaggerated allergic airway responses, suggesting that microbial host interactions play an important role in the development of allergic diseases. Probiotics modulate both the innate and adaptive inflammatory immune responses, often used as dietary supplements to provide health benefits in gastrointestinal disorders. Probiotics may serve as immunomodulators and activators of host defense pathways. Besides, oral probiotics can modulate the immune response in the respiratory system. Recently, studies in humans and animals have demonstrated the role of probiotic in RA and AA. To understand the characterization, microbiota, and the potential role of probiotics intervention of AA/AR, this review provides an overview of clinical features of AA and AR, probiotics for the prevention and treatment of AR, AA, changes in gut microbiota, and their mechanisms of action.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zenghui Jin
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geratology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
184
|
Cheema AS, Trevenen ML, Turlach BA, Furst AJ, Roman AS, Bode L, Gridneva Z, Lai CT, Stinson LF, Payne MS, Geddes DT. Exclusively Breastfed Infant Microbiota Develops over Time and Is Associated with Human Milk Oligosaccharide Intakes. Int J Mol Sci 2022; 23:2804. [PMID: 35269946 PMCID: PMC8910998 DOI: 10.3390/ijms23052804] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Temporal development of maternal and infant microbiomes during early life impacts short- and long-term infant health. This study aimed to characterize bacterial dynamics within maternal faecal, human milk (HM), infant oral, and infant faecal samples during the exclusive breastfeeding period and to document associations between human milk oligosaccharide (HMO) intakes and infant oral and faecal bacterial profiles. Maternal and infant samples (n = 10) were collected at 2−5, 30, 60, 90 and 120 days postpartum and the full-length 16S ribosomal RNA (rRNA) gene was sequenced. Nineteen HMOs were quantitated using high-performance liquid chromatography. Bacterial profiles were unique to each sample type and changed significantly over time, with a large degree of intra- and inter-individual variation in all sample types. Beta diversity was stable over time within infant faecal, maternal faecal and HM samples, however, the infant oral microbiota at day 2−5 significantly differed from all other time points (all p < 0.02). HMO concentrations and intakes significantly differed over time, and HMO intakes showed differential associations with taxa observed in infant oral and faecal samples. The direct clinical relevance of this, however, is unknown. Regardless, future studies should account for intakes of HMOs when modelling the impact of HM on infant growth, as it may have implications for infant microbiota development.
Collapse
Affiliation(s)
- Ali Sadiq Cheema
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Michelle Louise Trevenen
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia; (M.L.T.); (B.A.T.)
| | - Berwin Ashoka Turlach
- Centre for Applied Statistics, The University of Western Australia, Crawley, WA 6009, Australia; (M.L.T.); (B.A.T.)
| | - Annalee June Furst
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Ana Sophia Roman
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA 92093, USA; (A.J.F.); (A.S.R.); (L.B.)
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Zoya Gridneva
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Lisa Faye Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| | - Matthew Scott Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, WA 6008, Australia;
- Women and Infants Research Foundation, Subiaco, WA 6008, Australia
| | - Donna Tracy Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (A.S.C.); (Z.G.); (C.T.L.); (L.F.S.)
| |
Collapse
|
185
|
Gao X, Wang Z, Li X, Zhang X, Du S, Jia M, Hu D, Jia X, Cong B, Zhang Y, Ma C, Zhou S, Zhang J. A new Lactobacillus gasseri strain HMV18 inhibits the growth of pathogenic bacteria. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
186
|
Pandey U, Aich P. Postnatal intestinal mucosa and gut microbial composition develop hand in hand: A mouse study. Biomed J 2022; 46:100519. [PMID: 35306225 DOI: 10.1016/j.bj.2022.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/17/2022] [Accepted: 03/10/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND During the early postnatal life, gut microbiota development experiences dynamic changes in their structural and functional composition. The postnatal period is the critical window to develop a host defense mechanism. The maturation of intestinal mucosal barrier integrity is one of the essential defense mechanisms to prevent the entry of pathogens. However, the co-development of intestinal microbial colonization, formation of barrier integrity, and intestinal epithelial cell layer is not entirely understood. METHODS We studied the gut microbial composition and diversity using 16S rRNA marker gene-based sequencing in mice to understand postnatal age-dependent association kinetics between gut microbial and intestinal development. Next, we assessed the intestinal development by in vivo gut permeability assay, mRNA gene expression of different tight junction proteins and intestinal epithelial cell markers, goblet cells population, villus length, and cecal IgA quantification. RESULTS Our results showed a significant shift in gut microbial structural and functional composition from postnatal day 14 onwards with early life Proteobacteria abundance. Relative abundance of Verrucomicrobia was maximum at postnatal day 14 and showed a gradual decrease over time. We also observed an age-dependent biphasic pattern in barrier integrity improvement and differentiation of intestinal epithelial cells (IECs). A significant improvement in barrier integrity between days 1 and 7 showed the host factor contribution, while that beyond day 14 revealed an association with changes in microbiota composition. Our temporal correlation analysis associated Bacteroidetes phylum with the mucosal barrier formation during postnatal development. CONCLUSIONS The present study revealed the importance and interplay of host factors and the microbiome in gut development and intestinal mucosal homeostasis.
Collapse
|
187
|
Chen B, Huang H, Pan CQ. The role of gut microbiota in hepatitis B disease progression and treatment. J Viral Hepat 2022; 29:94-106. [PMID: 34415656 DOI: 10.1111/jvh.13595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 01/05/2023]
Abstract
Current therapeutic interventions can only suppress hepatitis B virus (HBV) replication or reduce complications without a cure. Therefore, further development of new treatment methods is critical for the global eradication of HBV. Accumulating evidence suggests that the liver and gut share an interconnected relationship referred to as the 'Gut-Liver Axis', where exchanges happen bi-directionally. The gut itself is the host to a unique microbiota profile which has metabolic, immunological, neurological and nutritional functions. Gut microbiota is not only constantly intersecting with the liver but also associated with hepatic injury when dysbiosis occurs. In recent years, there has been increased interest in gut microbiota and its implications on liver disease treatment. Progress has been made in understanding the complex relationship between chronic hepatitis B (CHB) and gut microbiota. New investigative techniques such as colony-free sequencing enabled new perspectives into this field. Mouse models and human studies revealed that HBV infection is associated with significant alteration of gut microbiota, which differ depending on the stage of CHB disease progression. Different mechanisms of the hepatic injury from gut microbiota dysbiosis have also been proposed based on findings of increased intestinal permeability to toxins, disruption of normal bacterial metabolism, and colonization of the gut by oral microbiota. New treatment methods targeting gut microbiota in CHB, such as probiotics and faecal microbiota transplant, have also gained promising results in recent years. The current review recapitulated the most recent investigations into the relationship between gut microbiota and CHB to provide research directions towards the new therapeutic target of CHB.
Collapse
Affiliation(s)
- Bryan Chen
- University of California, Los Angeles, California, USA
| | - Harry Huang
- Stony Brook University, Stony Brook, New York, USA
| | - Calvin Q Pan
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.,Division of Gastroenterology and Hepatology, Department of Medicine, NYU Langone Health, NYU School of Medicine, New York, New York, USA
| |
Collapse
|
188
|
The Microbiota-Gut Axis in Premature Infants: Physio-Pathological Implications. Cells 2022; 11:cells11030379. [PMID: 35159189 PMCID: PMC8834399 DOI: 10.3390/cells11030379] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Intriguing evidence is emerging in regard to the influence of gut microbiota composition and function on host health from the very early stages of life. The development of the saprophytic microflora is conditioned by several factors in infants, and peculiarities have been found for babies born prematurely. This population is particularly exposed to a high risk of infection, postnatal antibiotic treatment, feeding difficulties and neurodevelopmental disabilities. To date, there is still a wide gap in understanding all the determinants and the mechanism behind microbiota disruption and its influence in the development of the most common complications of premature infants. A large body of evidence has emerged during the last decades showing the existence of a bidirectional communication axis involving the gut microbiota, the gut and the brain, defined as the microbiota–gut–brain axis. In this context, given that very few data are available to demonstrate the correlation between microbiota dysbiosis and neurodevelopmental disorders in preterm infants, increasing interest has arisen to better understand the impact of the microbiota–gut–brain axis on the clinical outcomes of premature infants and to clarify how this may lead to alternative preventive, diagnostic and therapeutic strategies. In this review, we explored the current evidence regarding microbiota development in premature infants, focusing on the effects of delivery mode, type of feeding, environmental factors and possible influence of the microbiota–gut–brain axis on preterm clinical outcomes during their hospital stay and on their health status later in life.
Collapse
|
189
|
Sprenger N, Tytgat HL, Binia A, Austin S, Singhal A. Biology of human milk oligosaccharides: from Basic Science to Clinical Evidence. J Hum Nutr Diet 2022; 35:280-299. [PMID: 35040200 PMCID: PMC9304252 DOI: 10.1111/jhn.12990] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Human milk oligosaccharides (HMOs) have been researched by scientists for over 100 years, driven by the substantial evidence for the nutritional and health benefits of mother's milk. Yet research has truly bloomed during the last decade, thanks to the progress in biotechnology, which allowed the production of large amounts of bona fide HMOs. The availability of HMOs has been particularly crucial for the renewed interest in HMO research because of the low abundance or even absence of HMOs in farmed animal milk. This interest is reflected in the increasing number of original research publications and reviews on HMOs. Here, we provide an overview and critical discussion on structure function relations of HMOs that highlight why they are such interesting and important components of human milk. Clinical observations in breastfed infants backed by basic research from animal models provide guidance as to what physiological roles for HMOs are to be expected. From an evidence-based nutrition viewpoint, we discuss the current data supporting clinical relevance of specific HMOs based on randomized placebo controlled clinical intervention trials in formula-fed infants. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Hanne Lp Tytgat
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Aristea Binia
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sean Austin
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Atul Singhal
- Institute of Child Health, University College London, London, WC1N 1EH, United Kingdom
| |
Collapse
|
190
|
Diaz DMM, Aguirre MDCC, Escalera ALR, Gutiérrez MTT, Robles IO, Guzmán MJM, Díaz ALG, Peña MCG, Alvarado-Nájera AN, Domínguez IG, Villavicencio-Bautista JC, Rodríguez AAH, Marín-García R, González FJA, Wong AC, Guerra EG, Castañeda RD, Aguilar CAP, Zúñiga-Macías LP, Guerra JMA. Histologic characterization and risk factors for persistent albuminuria in adolescents in a region of highly prevalent end-stage renal failure of unknown origin. Clin Kidney J 2022; 15:1300-1311. [PMID: 35756733 PMCID: PMC9217647 DOI: 10.1093/ckj/sfac018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
End-stage renal failure of unknown origin (ESRD-UO) is a public health problem in Mexico and many regions of the world. The prevalence of ESRD-UO in Aguascalientes Mexico is one of the highest worldwide, particularly in adults between 20 and 40 years of age.
Aim
To screen adolescents for chronic kidney disease (CKD), identify risk factors, and histologically characterize adolescents with persistent albuminuria (pACR).
Methodology
This is a cross-sectional, observational, and comparative study of adolescents in whom serum creatinine and the albumin creatinine ratio (ACR) were determined when screening for CKD. A clinical evaluation and risk factor survey were conducted. Patients with an abnormal ACR (≥ 30 mg/gr) or a low glomerular filtration rate (GFR) (≤75 ml/min/1.73 m2) were reevaluated and a renal ultrasound was obtained (rUS). A kidney biopsy was performed in patients with pACR.
Results
Five-hundred and thirteen (513) students were included, 19 had pACR and 494 were controls. The prevalence of pACR was 3.7% (95%CI 2.1 – 5.3). Only one patient had a decreased GRF. None of the patients with pACR had anatomical abnormalities of the urinary tract by rUS. Patients with pACR had a decreased total renal volume in comparison with the control group (150 vs 195 ml/m2, p <0.01). Eighteen (18) kidney biopsies were performed, 72% had glomerulomegaly, and only one patient had mild fibrosis. Podocyte abnormalities were evident on electron microscopy: partial fusion (100%), microvillous degeneration (80%), and increased organelles (60%). Risk factors for pACR were: homestead proximity to maize crops, the use of pesticides at the father´s workplace, a family history of CKD, and blood pressure abnormalities. The body mass index and breastfeeding were protective factors.
Conclusions
The prevalence of pACR in adolescents in Aguascalientes is high, and histologic compromise is characterized by podocyte injury in the absence of fibrosis. The renal volume of pACR patients was decreased, suggesting oligonephronia. Exposure to environmental toxins such as pesticides, even prenatally, may be responsible for this pathological entity. Screening programs in adolescents by determining ACR are necessary in our milieu.
Collapse
Affiliation(s)
| | - Myriam del Carmen Corrales Aguirre
- Department of Nephrology, Hospital Centenario Miguel Hidalgo, Mexico
- Department of Pediatrics, Hospital Centenario Miguel Hidalgo, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | | - Alfredo Chew Wong
- Department of Nephrology, Hospital Centenario Miguel Hidalgo, Mexico
| | | | - Rodolfo Delgadillo Castañeda
- Department of Nephrology, Hospital Centenario Miguel Hidalgo, Mexico
- Department of Pediatrics, Hospital Centenario Miguel Hidalgo, Mexico
| | | | | | | |
Collapse
|
191
|
Yamamoto S, Murata T. [The effect of bioactive substances derived from gut bacteria on the development of allergic diseases]. Nihon Yakurigaku Zasshi 2022; 157:85. [PMID: 34980817 DOI: 10.1254/fpj.21088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
192
|
Ahn JR, Lee SH, Kim B, Nam MH, Ahn YK, Park YM, Jeong SM, Park MJ, Song KB, Lee SY, Hong SJ. Ruminococcus gnavus ameliorates atopic dermatitis by enhancing Treg cell and metabolites in BALB/c mice. Pediatr Allergy Immunol 2022; 33:e13678. [PMID: 34633714 DOI: 10.1111/pai.13678] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ruminococcus gnavus (R. gnavus) are mucin-degrading gut bacteria that play a key role in the early colonization of the gut by serving as endogenous sources of nutrients. They can also influence immune development. We had previously reported a lower abundance of R. gnavus in infants with atopic dermatitis (AD) compared with that in healthy subjects. However, the underlying mechanisms remain unclear. In this study, we investigated the effect of orally administered R. gnavus on antibiotic treatment-induced gut dysbiosis (and the underlying mechanism) in a mouse model of AD. METHODS Four-week-old female BALB/C mice were administered antibiotic cocktails for 2 weeks. R. gnavus was orally administered throughout the study duration. At 6 weeks of age, AD was induced by epidermal sensitization with ovalbumin. AD phenotypes and systemic and gut immune responses were investigated. RESULTS Orally administered R. gnavus significantly reduced AD-associated parameters (i.e., transepidermal water loss, clinical score, total serum immunoglobulin (Ig) E level, OVA-specific IgE level, and skin inflammation). R. gnavus treatment also resulted in significant downregulation of T helper 2-related cytokine mRNA and upregulation of interleukin (IL)-10 and Foxp3 in the skin. The population of CD4+ FOXP3+ T cells in mesenteric- and skin-draining lymph nodes and butyrate levels in the cecum increased in R. gnavus-administered AD mice. CONCLUSIONS Immune modulation by orally administered R. gnavus may alleviate AD symptoms through the enhancement of regulatory T-cell counts and short-chain fatty acids production in AD mice.
Collapse
Affiliation(s)
- Jae-Rin Ahn
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Hwa Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Byunghyun Kim
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Myung Hee Nam
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Yoon Kyung Ahn
- Korea Basic Science Institute, Western Seoul Center, Seoul, Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seon-Mi Jeong
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Jee Park
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kun Baek Song
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
193
|
Aganetti MA, Cruz CS, Galvão I, Engels DF, Ricci MF, Vieira AT. The Gut Microbiota and Immunopathophysiology. COMPREHENSIVE PHARMACOLOGY 2022:492-514. [DOI: 10.1016/b978-0-12-820472-6.00128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
194
|
de Cuevillas B, Milagro FI, Tur JA, Gil-Campos M, de Miguel-Etayo P, Martínez JA, Navas-Carretero S. Fecal microbiota relationships with childhood obesity: A scoping comprehensive review. Obes Rev 2022; 23 Suppl 1:e13394. [PMID: 34913242 DOI: 10.1111/obr.13394] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022]
Abstract
Childhood obesity is a costly burden in most regions with relevant and adverse long-term health consequences in adult life. Several studies have associated excessive body weight with a specific profile of gut microbiota. Different factors related to fecal microorganism abundance seem to contribute to childhood obesity, such as gestational weight gain, perinatal diet, antibiotic administration to the mother and/or child, birth delivery, and feeding patterns, among others. This review reports and discusses diverse factors that affect the infant intestinal microbiota with putative or possible implications on the increase of the obesity childhood rates as well as microbiota shifts associated with excessive body weight in children.
Collapse
Affiliation(s)
- Begoña de Cuevillas
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Fermín I Milagro
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| | - Josep A Tur
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS & IDISBA, Palma de Mallorca, Spain
| | - Mercedes Gil-Campos
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Metabolism and Investigation Unit, Reina Sofia University Hospital, Maimónides Institute of Biomedicine Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Pilar de Miguel-Etayo
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza. Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - J Alfredo Martínez
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
- Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food. CSIC-UAM, Madrid, Spain
| | - Santiago Navas-Carretero
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| |
Collapse
|
195
|
Vitamin D Supplementation in Exclusively Breastfed Infants Is Associated with Alterations in the Fecal Microbiome. Nutrients 2022; 14:nu14010202. [PMID: 35011077 PMCID: PMC8747039 DOI: 10.3390/nu14010202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Breastfeeding and introduction of solid food are the two major components of infant feeding practices that influence gut microbiota composition in early infancy. However, it is unclear whether additional factors influence the microbiota of infants either exclusively breastfed or not breastfed. We obtained 194 fecal samples from infants at 3–9 months of age, extracted DNA, and sequenced the V4 region of the 16S rRNA gene. Feeding practices and clinical information were collected by questionnaire and abstraction of birth certificates. The gut microbiota of infants who were exclusively breastfed displayed significantly lower Shannon diversity (p-adjust < 0.001) and different gut microbiota composition compared to infants who were not breastfed (p-value = 0.001). Among the exclusively breastfed infants, recipients of vitamin D supplements displayed significantly lower Shannon diversity (p-adjust = 0.007), and different gut microbiota composition structure than non-supplemented, breastfed infants (p-value = 0.02). MaAslin analysis identified microbial taxa that associated with breastfeeding and vitamin D supplementation. Breastfeeding and infant vitamin D supplement intake play an important role in shaping infant gut microbiota.
Collapse
|
196
|
Spontaneous preterm delivery is reflected in both early neonatal and maternal gut microbiota. Pediatr Res 2022; 91:1804-1811. [PMID: 34349229 PMCID: PMC9270225 DOI: 10.1038/s41390-021-01663-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/22/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Aberrant gut microbiota composition in preterm neonates is linked to adverse health consequences. Little is known about the impact of perinatal factors or maternal gut microbiota on initial preterm gut colonization. METHODS Fecal samples were collected from 55 preterm neonates (<35 gestational weeks), 51 mothers, and 25 full-term neonates during the first 3-4 postpartum days. Gut microbiota composition was assessed using 16S ribosomal RNA gene sequencing. RESULTS Preterm neonates exhibited significantly lower gut microbiota alpha diversity and distinct beta diversity clustering compared to term neonates. Spontaneous preterm birth was associated with distinct initial gut microbiota beta diversity as compared to iatrogenic delivery. Gestational age or delivery mode had no impact on the preterm gut microbiota composition. The cause of preterm delivery was also reflected in the maternal gut microbiota composition. The contribution of maternal gut microbiota to initial preterm gut colonization was more pronounced after spontaneous delivery than iatrogenic delivery and not dependent on delivery mode. CONCLUSIONS The initial preterm gut microbiota is distinct from term microbiota. Spontaneous preterm birth is reflected in the early neonatal and maternal gut microbiota. Transmission of gut microbes from mother to neonate is determined by spontaneous preterm delivery, but not by mode of birth. IMPACT The initial gut microbiota in preterm neonates is distinct from those born full term. Spontaneous preterm birth is associated with changes in the gut microbiota composition of both preterm neonates and their mothers. The contribution of the maternal gut microbiota to initial neonatal gut colonization was more pronounced after spontaneous preterm delivery as compared to iatrogenic preterm delivery and not dependent on delivery mode. Our study provides new evidence regarding the early gut colonization patterns in preterm infants. Altered preterm gut microbiota has been linked to adverse health consequences and may provide a target for early intervention.
Collapse
|
197
|
Mundula T, Russo E, Curini L, Giudici F, Piccioni A, Franceschi F, Amedei A. Chronic Systemic Low-Grade Inflammation and Modern Lifestyle: The Dark Role of Gut Microbiota on Related Diseases with a Focus on COVID-19 Pandemic. Curr Med Chem 2022; 29:5370-5396. [PMID: 35524667 DOI: 10.2174/0929867329666220430131018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022]
Abstract
Inflammation is a physiological, beneficial, and auto-limiting response of the host to alarming stimuli. Conversely, a chronic systemic low-grade inflammation (CSLGI), known as a long-time persisting condition, causes damage to the organs and host tissues, representing a major risk for chronic diseases. Currently, a high global incidence of chronic inflammatory diseases is observed, often linked to the lifestyle-related changes that occurred in the last decade. The main lifestyle-related factors are proinflammatory diet, psychological stress, tobacco smoking, alcohol abuse, physical inactivity, and indoor living and working with its related consequences such as indoor pollution, artificial light exposure, and low vitamin D production. Recent scientific evidence found that gut microbiota (GM) has a main role in shaping the host's health, particularly as CSLGI mediator. Based on the lastest discoveries regarding the remarkable GM activity, in this manuscript we focus on the elements of actual lifestyle that influence the composition and function of the intestinal microbial community in order to elicit the CSLGI and its correlated pathologies. In this scenario, we provide a broad review of the interplay between modern lifestyle, GM, and CSLGI with a special focus on the COVID symptoms and emerging long-COVID syndrome.
Collapse
Affiliation(s)
- Tiziana Mundula
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, University of Florence
| | - Lavinia Curini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Giudici
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Piccioni
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Franceschi
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
198
|
de Goffau MC, Jallow AT, Sanyang C, Prentice AM, Meagher N, Price DJ, Revill PA, Parkhill J, Pereira DIA, Wagner J. Gut microbiomes from Gambian infants reveal the development of a non-industrialized Prevotella-based trophic network. Nat Microbiol 2022; 7:132-144. [PMID: 34972822 PMCID: PMC8727306 DOI: 10.1038/s41564-021-01023-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Distinct bacterial trophic networks exist in the gut microbiota of individuals in industrialized and non-industrialized countries. In particular, non-industrialized gut microbiomes tend to be enriched with Prevotella species. To study the development of these Prevotella-rich compositions, we investigated the gut microbiota of children aged between 7 and 37 months living in rural Gambia (616 children, 1,389 stool samples, stratified by 3-month age groups). These infants, who typically eat a high-fibre, low-protein diet, were part of a double-blind, randomized iron intervention trial (NCT02941081) and here we report the secondary outcome. We found that child age was the largest discriminating factor between samples and that anthropometric indices (collection time points, season, geographic collection site, and iron supplementation) did not significantly influence the gut microbiome. Prevotella copri, Faecalibacterium prausnitzii and Prevotella stercorea were, on average, the most abundant species in these 1,389 samples (35%, 11% and 7%, respectively). Distinct bacterial trophic network clusters were identified, centred around either P. stercorea or F. prausnitzii and were found to develop steadily with age, whereas P. copri, independently of other species, rapidly became dominant after weaning. This dataset, set within a critical gut microbial developmental time frame, provides insights into the development of Prevotella-rich gut microbiomes, which are typically understudied and are underrepresented in western populations.
Collapse
Affiliation(s)
- Marcus C de Goffau
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, UK
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Amadou T Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Chilel Sanyang
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Andrew M Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Niamh Meagher
- Department of Infectious Diseases at the Doherty Institute for Infection & Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, The University of Melbourne, Melbourne, Australia
| | - David J Price
- Department of Infectious Diseases at the Doherty Institute for Infection & Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, The University of Melbourne, Melbourne, Australia
| | - Peter A Revill
- Victorian Infectious Disease Reference Laboratory, the Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Dora I A Pereira
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Josef Wagner
- Parasites and Microbes, Wellcome Sanger Institute, Cambridge, UK.
- Victorian Infectious Disease Reference Laboratory, the Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia.
| |
Collapse
|
199
|
Lee MJ, Park YM, Kim B, Tae IH, Kim NE, Pranata M, Kim T, Won S, Kang NJ, Lee YK, Lee DW, Nam MH, Hong SJ, Kim BS. Disordered development of gut microbiome interferes with the establishment of the gut ecosystem during early childhood with atopic dermatitis. Gut Microbes 2022; 14:2068366. [PMID: 35485368 PMCID: PMC9067516 DOI: 10.1080/19490976.2022.2068366] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
The gut microbiome influences the development of allergic diseases during early childhood. However, there is a lack of comprehensive understanding of microbiome-host crosstalk. Here, we analyzed the influence of gut microbiome dynamics in early childhood on atopic dermatitis (AD) and the potential interactions between host and microbiome that control this homeostasis. We analyzed the gut microbiome in 346 fecal samples (6-36 months; 112 non-AD, 110 mild AD, and 124 moderate to severe AD) from the Longitudinal Cohort for Childhood Origin of Asthma and Allergic Disease birth cohort. The microbiome-host interactions were analyzed in animal and in vitro cell assays. Although the gut microbiome maturated with age in both AD and non-AD groups, its development was disordered in the AD group. Disordered colonization of short-chain fatty acids (SCFA) producers along with age led to abnormal SCFA production and increased IgE levels. A butyrate deficiency and downregulation of GPR109A and PPAR-γ genes were detected in AD-induced mice. Insufficient butyrate decreases the oxygen consumption rate of host cells, which can release oxygen to the gut and perturb the gut microbiome. The disordered gut microbiome development could aggravate balanced microbiome-host interactions, including immune responses during early childhood with AD.
Collapse
Affiliation(s)
- Min-Jung Lee
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon, Republic of Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Byunghyun Kim
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - in Hwan Tae
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Nam-Eun Kim
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Marina Pranata
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan, Republic of Korea
| | - Taewon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Nam Joo Kang
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
- Department of Integrative Biology, Kyungpook National University, Daegu, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Myung Hee Nam
- Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Kim
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon, Republic of Korea
- The Korean Institute of Nutrition, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
200
|
AboNahas HH, Darwish AMG, Abd EL-kareem HF, AboNahas YH, Mansour SA, Korra YH, Sayyed RZ, Abdel-Azeem AM, Saied EM. Trust Your Gut: The Human Gut Microbiome in Health and Disease. MICROBIOME-GUT-BRAIN AXIS 2022:53-96. [DOI: 10.1007/978-981-16-1626-6_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|