1
|
Al-Beltagi M. Human milk oligosaccharide secretion dynamics during breastfeeding and its antimicrobial role: A systematic review. World J Clin Pediatr 2025; 14. [DOI: 10.5409/wjcp.v14.i2.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND
Human milk oligosaccharides (HMOs) are bioactive components of breast milk with diverse health benefits, including shaping the gut microbiota, modulating the immune system, and protecting against infections. HMOs exhibit dynamic secretion patterns during lactation, influenced by maternal genetics and environmental factors. Their direct and indirect antimicrobial properties have garnered significant research interest. However, a comprehensive understanding of the secretion dynamics of HMOs and their correlation with antimicrobial efficacy remains underexplored.
AIM
To synthesize current evidence on the secretion dynamics of HMOs during lactation and evaluate their antimicrobial roles against bacterial, viral, and protozoal pathogens.
METHODS
A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library focused on studies investigating natural and synthetic HMOs, their secretion dynamics, and antimicrobial properties. Studies involving human, animal, and in vitro models were included. Data on HMO composition, temporal secretion patterns, and mechanisms of antimicrobial action were extracted. Quality assessment was performed using validated tools appropriate for study design.
RESULTS
A total of 44 studies were included, encompassing human, animal, and in vitro research. HMOs exhibited dynamic secretion patterns, with 2′-fucosyllactose (2′-FL) and lacto-N-tetraose peaking in early lactation and declining over time, while 3-fucosyllactose (3-FL) increased during later stages. HMOs demonstrated significant antimicrobial properties through pathogen adhesion inhibition, biofilm disruption, and enzymatic activity impairment. Synthetic HMOs, including bioengineered 2′-FL and 3-FL, were structurally and functionally comparable to natural HMOs, effectively inhibiting pathogens such as Pseudomonas aeruginosa , Escherichia coli , and Campylobacter jejuni . Additionally, HMOs exhibited synergistic effects with antibiotics, enhancing their efficacy against resistant pathogens.
CONCLUSION
HMOs are vital in antimicrobial defense, supporting infant health by targeting various pathogens. Both natural and synthetic HMOs hold significant potential for therapeutic applications, particularly in infant nutrition and as adjuncts to antibiotics. Further research, including clinical trials, is essential to address gaps in knowledge, validate findings, and explore the broader applicability of HMOs in improving maternal and neonatal health.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Paediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
2
|
Tang H, Li W, Xu Y, Zhou Y, Hamblin MR, Wen X. Gut microbiota modulation: a key determinant of atopic dermatitis susceptibility in children. Front Microbiol 2025; 16:1549895. [PMID: 40356648 PMCID: PMC12066585 DOI: 10.3389/fmicb.2025.1549895] [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: 12/22/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Atopic dermatitis is a chronic inflammatory skin condition with a higher incidence rate among children. In recent years, the role of the gut microbiota in the pathogenesis of atopic dermatitis has garnered increasing attention. This review systematically delineates the research advancements in the structural characteristics of the gut microbiota in children with atopic dermatitis and its influencing factors. Studies have revealed significant differences in the gut microbiota structure between children with atopic dermatitis and healthy controls, characterized by a reduction in microbial diversity, a decrease in beneficial bacteria, and an increase in harmful bacteria. Dietary patterns, environmental factors, birth patterns, antibiotic use, and gestational diabetes mellitus are factors could impact the gut microbiota hence influencing the susceptibility of children to atopic dermatitis. Moreover, this review explores the interplay between the gut microbiota and the immune system in atopic dermatitis, with the potential to inform more effective probiotic treatment strategies for children with atopic dermatitis.
Collapse
Affiliation(s)
- Huimiao Tang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxin Li
- The International Department of Chengdu Shude High School, Chengdu, China
| | - Yidan Xu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanjun Zhou
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Hu P, Sun J, Gao R, Li K, Liu J, Pan X, Jin Z, Mao Y, Yang J, Yu R, Qi C. Harnessing the power of breast milk: how Lactiplantibacillus plantarum FN029 from rural western China mitigates severe atopic dermatitis in mice through retinol metabolism activation. Food Funct 2025; 16:2230-2246. [PMID: 39912208 DOI: 10.1039/d4fo04300f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Tongwei and Wuxi represent a rural county in western China and an industrialized city in the east, respectively. The study compared breast milk and the corresponding infant gut microbiota from 35 healthy mothers in Tongwei and 28 in Wuxi, uncovering significant differences in microbial alpha and beta diversity. A unique strain, Lactiplantibacillus plantarum FN029, characteristically transmitted from breast milk to the infant gut in Tongwei, was identified. Oral administration of FN029 to weaned BALB/c mice significantly alleviated atopic dermatitis severity caused by calcipotriol and ovalbumin. This reduction was paralleled by a decrease in mast cells and eosinophils in ear tissue and reduced levels of IL-4, IL-12, IL-33, IFN-γ, the IL-4/IFN-γ ratio, and IgE in plasma, along with an upsurge in regulatory T cells in the spleen. RNA sequencing revealed that FN029 activated the retinol metabolism pathway and the Wnt signaling pathway, enhancing immature dendritic cells and regulatory T cells. Metabolomics analysis indicated an increase in retinyl beta-glucuronide, a biomarker of vitamin A reserves. The mRNA expression of retinol-metabolizing enzymes was inversely related to the IL-4/IFN-γ ratio. FN029 also altered ileum microbiota without a direct link to atopic dermatitis improvement. In conclusion, L. plantarum FN029, a probiotic from Tongwei breast milk, fostered T regulatory cell conversion and immune balance by activating the retinol pathway, thereby improving severe atopic dermatitis in mice.
Collapse
Affiliation(s)
- Pengyue Hu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Ruijuan Gao
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Kexin Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Jiayi Liu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Xiaonan Pan
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Zilu Jin
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| | - Yuejian Mao
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy(Group) Co. Ltd, Hohhot, Inner Mongolia, China
| | - Jing Yang
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy(Group) Co. Ltd, Hohhot, Inner Mongolia, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Jiangnan University, Wuxi 214002, PR China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.
| |
Collapse
|
4
|
Hoskinson C, Petersen C, Turvey SE. How the early life microbiome shapes immune programming in childhood asthma and allergies. Mucosal Immunol 2025; 18:26-35. [PMID: 39675725 DOI: 10.1016/j.mucimm.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
Despite advances in our understanding of their diagnosis and treatment, pediatric allergies impose substantial burdens on affected children, families, and healthcare systems. Further, the prevalence of allergic diseases has dramatically increased over the past half-century, leading to additional concerns and concerted efforts to identify the origins, potential predictors and preventions, and therapies of allergic diseases. Together with the increase in allergic diseases, changes in lifestyle and early-life environmental influences have corresponded with changes in colonization patterns of the infant gut microbiome. The gut microbiome plays a key role in developing the immune system, thus greatly influencing the development of allergic disease. In this review, we specifically highlight the importance of the proper maturation and composition of the gut microbiome as an essential step in healthy child development or disease progression. By exploring the intertwined development of the immune system and microbiome across pediatric allergic diseases, we provide insights into potential novel strategies for their prevention and management.
Collapse
Affiliation(s)
- Courtney Hoskinson
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Charisse Petersen
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Zhao J, Guo Y, Jiang Q, Lan H, Hung WL, Lynch B. Bifidobacterium longum subsp. infantis YLGB-1496-Toxicological evaluation. J Appl Toxicol 2025; 45:230-244. [PMID: 39252460 DOI: 10.1002/jat.4688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/11/2024]
Abstract
Bifidobacterium infantis YLGB-1496, originally isolated from breast milk from a Taiwanese mother, is under study for use as a probiotic. As part of safety assessment, an Ames, in vivo mouse micronucleus, and in vivo mouse spermatocyte chromosome aberration assay were conducted along with a 13-week oral rat toxicity study. B. infantis YLGB-1496 had no activity in any of the genotoxicity assays. Administration of the bacteria to Sprague-Dawley rats at doses ranging from 0 to 1.5 g/kg bw/day had no treatment-related effects on any of the endpoints measured. There appear to be no concerns for translocation or pathogenicity of B. infantis YLGB-1496 based on extensive experience with the species in general. The results of the current investigations support potential use of B. infantis YLGB-1496 as a probiotic in infant formula.
Collapse
Affiliation(s)
- Jian Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yueyi Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qiuyue Jiang
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Barry Lynch
- Intertek Health Sciences Inc., Mississauga, ON, Canada
| |
Collapse
|
6
|
Scher JU, Nayak R, Clemente JC. Microbiome research in autoimmune and immune-mediated inflammatory diseases: lessons, advances and unmet needs. Ann Rheum Dis 2025; 84:9-13. [PMID: 39874238 PMCID: PMC11822223 DOI: 10.1136/ard-2024-225735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
The increasing prevalence of autoimmune and immune-mediated diseases (AIMDs) underscores the need to understand environmental factors that contribute to their pathogenesis, with the microbiome emerging as a key player. Despite significant advancements in understanding how the microbiome influences physiological and inflammatory responses, translating these findings into clinical practice remains challenging. This viewpoint reviews the progress and obstacles in microbiome research related to AIMDs, examining molecular techniques that enhance our understanding of microbial contributions to disease. We discuss significant discoveries linking specific taxa and metabolites to diseases such as rheumatoid arthritis, systemic lupus erythematosus and spondyloarthritis, highlighting the role of gut dysbiosis and host-microbiome interactions. Furthermore, we explore the potential of microbiome-based therapeutics, including faecal microbiota transplantation and pharmacomicrobiomics, while addressing the challenges of identifying robust microbial targets. We advocate for integrative, transdisease studies and emphasise the need for diverse cohort research to generalise findings across populations. Understanding the microbiome's role in AIMDs will pave the way for personalised medicine and innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jose U Scher
- Department of Medicine, NYU Psoriatic Arthritis Center, and NYU Colton Center for Autoimmunity, New York University School of Medicine, New York, New York, USA.
| | - Renuka Nayak
- University of California San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, San Francisco, California, USA. https://x.com/RNayak
| | - Jose C Clemente
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Dai DLY, Petersen C, Turvey SE. Reduce, reinforce, and replenish: safeguarding the early-life microbiota to reduce intergenerational health disparities. Front Public Health 2024; 12:1455503. [PMID: 39507672 PMCID: PMC11537995 DOI: 10.3389/fpubh.2024.1455503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Socioeconomic (SE) disparity and health inequity are closely intertwined and associated with cross-generational increases in the rates of multiple chronic non-communicable diseases (NCDs) in North America and beyond. Coinciding with this social trend is an observed loss of biodiversity within the community of colonizing microbes that live in and on our bodies. Researchers have rightfully pointed to the microbiota as a key modifiable factor with the potential to ease existing health inequities. Although a number of studies have connected the adult microbiome to socioeconomic determinants and health outcomes, few studies have investigated the role of the infant microbiome in perpetuating these outcomes across generations. It is an essential and important question as the infant microbiota is highly sensitive to external forces, and observed shifts during this critical window often portend long-term outcomes of health and disease. While this is often studied in the context of direct modulators, such as delivery mode, family size, antibiotic exposure, and breastfeeding, many of these factors are tied to underlying socioeconomic and/or cross-generational factors. Exploring cross-generational socioeconomic and health inequities through the lens of the infant microbiome may provide valuable avenues to break these intergenerational cycles. In this review, we will focus on the impact of social inequality in infant microbiome development and discuss the benefits of prioritizing and restoring early-life microbiota maturation for reducing intergenerational health disparities.
Collapse
Affiliation(s)
| | | | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Mousavian AH, Zare Garizi F, Ghoreshi B, Ketabi S, Eslami S, Ejtahed HS, Qorbani M. The association of infant and mother gut microbiomes with development of allergic diseases in children: a systematic review. J Asthma 2024; 61:1121-1135. [PMID: 38506489 DOI: 10.1080/02770903.2024.2332921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
OBJECTIVE It is believed that gut microbiota alteration leads to both intestinal and non-intestinal diseases in children. Since infants inherit maternal microbiota during pregnancy and lactation, recent studies suggest that changes in maternal microbiota can cause immune disorders as well. This systematic review was designed to assess the association between the child and mother's gut microbiome and allergy development in childhood. DATA SOURCES In this systematic review, international databases including PubMed, Scopus, and ISI/WOS were searched until January 2023 to identify relevant studies. STUDY SELECTIONS Observational studies that analyzed infant or maternal stool microbiome and their association with allergy development in children were included in this study. Data extraction and quality assessment of the included studies were independently conducted by two researchers. RESULTS Of the 1694 papers evaluated, 21 studies examined neonate gut microbiome by analyzing stool samples and six studies examined maternal gut microbiota. A total of 5319 participants were included in this study. Asthma followed by eczema and dermatitis were the most common allergy disorders among children. Urbanization caused a lack of diversity in the bacterial microbiota as well as lower levels of Bifidobacterium and Lachnospira associated with a higher risk of allergy. In contrast, higher levels of Roseburia and Flavonifractor were associated with lower allergy risk. CONCLUSIONS This systematic review shows that gut microbiota may be associated with allergy development. Further studies are required to provide a definitive answer.
Collapse
Affiliation(s)
- Amir-Hossein Mousavian
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Zare Garizi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Behnaz Ghoreshi
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Siavash Ketabi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
9
|
Tripp P, Davis EC, Gurung M, Rosa F, Bode L, Fox R, LeRoith T, Simecka C, Seppo AE, Järvinen KM, Yeruva L. Infant Microbiota Communities and Human Milk Oligosaccharide Supplementation Independently and Synergistically Shape Metabolite Production and Immune Responses in Healthy Mice. J Nutr 2024; 154:2871-2886. [PMID: 39069270 PMCID: PMC11393170 DOI: 10.1016/j.tjnut.2024.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Multiple studies have demonstrated associations between the early-life gut microbiome and incidence of inflammatory and autoimmune disease in childhood. Although microbial colonization is necessary for proper immune education, it is not well understood at a mechanistic level how specific communities of bacteria promote immune maturation or drive immune dysfunction in infancy. OBJECTIVES In this study, we aimed to assess whether infant microbial communities with different overall structures differentially influence immune and gastrointestinal development in healthy mice. METHODS Germ-free mice were inoculated with fecal slurries from Bifidobacterium longum subspecies infantis positive (BIP) or B. longum subspecies infantis negative (BIN) breastfed infants; half of the mice in each group were also supplemented with a pool of human milk oligosaccharides (HMOs) for 14 d. Cecal microbiome composition and metabolite production, systemic and mucosal immune outcomes, and intestinal morphology were assessed at the end of the study. RESULTS The results showed that inoculation with a BIP microbiome results in a remarkably distinct microbial community characterized by higher relative abundances of cecal Clostridium senu stricto, Ruminococcus gnavus, Cellulosilyticum sp., and Erysipelatoclostridium sp. The BIP microbiome produced 2-fold higher concentrations of cecal butyrate, promoted branched short-chain fatty acid (SCFA) production, and further modulated serotonin, kynurenine, and indole metabolism relative to BIN mice. Further, the BIP microbiome increased the proportions of innate and adaptive immune cells in spleen, while HMO supplementation increased proliferation of mesenteric lymph node cells to phorbol myristate acetate and lipopolysaccharide and increased serum IgA and IgG concentrations. CONCLUSIONS Different microbiome compositions and HMO supplementation can modulate SCFA and tryptophan metabolism and innate and adaptive immunity in young, healthy mice, with potentially important implications for early childhood health.
Collapse
Affiliation(s)
- Patricia Tripp
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Manoj Gurung
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Fernanda Rosa
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States; Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Renee Fox
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Tanya LeRoith
- Department of Biomedical Sciences & Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Christy Simecka
- Division of Laboratory Animal Medicine University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY, United States; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| | - Laxmi Yeruva
- USDA-ARS, SEA, Microbiome and Metabolism Research Unit, Arkansas Children's Nutrition Center, Little Rock, AR, United States.
| |
Collapse
|
10
|
Davis EC, Monaco CL, Insel R, Järvinen KM. Gut microbiome in the first 1000 days and risk for childhood food allergy. Ann Allergy Asthma Immunol 2024; 133:252-261. [PMID: 38494114 PMCID: PMC11344696 DOI: 10.1016/j.anai.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
OBJECTIVE To summarize recent data on the association between gut microbiome composition and food allergy (FA) in early childhood and highlight potential host-microbiome interactions that reinforce or abrogate oral tolerance. DATA SOURCES PubMed search of English-language articles related to FA, other atopic disease, and the gut microbiome in pregnancy and early childhood. STUDY SELECTIONS Human studies published after 2015 assessing the relationship between the gut bacteriome and virome in the first 2 years of life and FA or food sensitization development in early childhood were prioritized. Additional human studies conducted on the prenatal gut microbiome or other atopic diseases and preclinical studies are also discussed. RESULTS Children who developed FA harbored lower abundances of Bifidobacterium and Clostridia species and had a less mature microbiome during infancy. The early bacterial microbiome protects against FA through production of anti-inflammatory metabolites and induction of T regulatory cells and may also affect FA risk through a role in trained immunity. Infant enteric phage communities are related to childhood asthma development, though no data are available for FA. Maternal gut microbiome during pregnancy is associated with childhood FA risk, potentially through transplacental delivery of maternal bacterial metabolites, though human studies are lacking. CONCLUSION The maternal and infant microbiomes throughout the first 1000 days of life influence FA risk through a number of proposed mechanisms. Further large, longitudinal cohort studies using taxonomic, functional, and metabolomic analysis of the bacterial and viral microbiomes are needed to provide further insight on the host-microbe interactions underlying FA pathogenesis in childhood.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Cynthia L Monaco
- Division of Infectious Disease, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Richard Insel
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
11
|
Rothenberg-Lausell C, David E, Del Duca E, Da Rosa JC, Dahabreh D, Gómez-Arias PJ, Catlin E, Nandymazumdar M, Järvinen KM, Guttman-Yassky E. Skin tape-strips in old order Mennonite toddlers reveal upregulated barrier markers and low T-helper inflammatory tone. Allergy 2024; 79:2012-2015. [PMID: 38814244 PMCID: PMC11236509 DOI: 10.1111/all.16154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/04/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Affiliation(s)
- Camille Rothenberg-Lausell
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Eden David
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ester Del Duca
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Dermatology, University of La Sapienza, Rome, Italy
| | - Joel Correa Da Rosa
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dante Dahabreh
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pedro Jesús Gómez-Arias
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Dermatology and Immune-Mediated Inflammatory Skin Diseases Research Group, IMIBIC/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
| | - Elizabeth Catlin
- Department of Pediatrics, Division of Allergy and Immunology, Center for Food Allergy, University of Rochester School of Medicine, Rochester, New York
| | - Monali Nandymazumdar
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kirsi M. Järvinen
- Department of Pediatrics, Division of Allergy and Immunology, Center for Food Allergy, University of Rochester School of Medicine, Rochester, New York
| | - Emma Guttman-Yassky
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
12
|
Pizzarello CR, Nelson A, Verekhman I, Seppo AE, Järvinen KM. Human milk affects TLR4 activation and LPS-induced inflammatory cytokine expression in Caco-2 intestinal epithelial cells. Sci Rep 2024; 14:13448. [PMID: 38862662 PMCID: PMC11167050 DOI: 10.1038/s41598-024-64000-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Human milk (HM) components affect immune cell toll-like receptor 4 (TLR4) signaling. However, studies examining the immunomodulatory impacts of HM on TLR4 signaling in intestinal epithelial cells (IECs) are limited. This study utilized both a TLR4 reporter cell line and a Caco-2 IEC model to examine the effects of HM on lipopolysaccharide (LPS)-induced TLR4 activation and cytokine responses, respectively. Additionally, we performed fast protein liquid chromatography and mass spectrometry to identify a HM component that contributes to the effect of HM on LPS/TLR4 signaling. HM enhances LPS-induced TLR4 signaling as well as LPS-induced IEC gene expression of pro-inflammatory cytokines and negative regulators of NF-κB. Human serum albumin (HSA) present in HM contributes to these effects. HSA within HM synergizes with LPS to induce IEC gene expression of pro-inflammatory cytokines and negative regulators of NF-κB. Altogether, this study provides mechanistic evidence behind the immunomodulatory function of HM on IECs, which may contribute to an enhanced immune response in breast-fed neonates.
Collapse
Affiliation(s)
- Catherine R Pizzarello
- Division of Pediatric Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, 601 Elmwood Ave, Box 777, Rochester, NY, 14642, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ashley Nelson
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ilya Verekhman
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Antti E Seppo
- Division of Pediatric Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, 601 Elmwood Ave, Box 777, Rochester, NY, 14642, USA
| | - Kirsi M Järvinen
- Division of Pediatric Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, 601 Elmwood Ave, Box 777, Rochester, NY, 14642, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
13
|
Campo JJ, Seppo AE, Randall AZ, Pablo J, Hung C, Teng A, Shandling AD, Truong J, Oberai A, Miller J, Iqbal NT, Peñataro Yori P, Kukkonen AK, Kuitunen M, Guterman LB, Morris SK, Pell LG, Al Mahmud A, Ramakrishan G, Heinz E, Kirkpatrick BD, Faruque AS, Haque R, Looney RJ, Kosek MN, Savilahti E, Omer SB, Roth DE, Petri WA, Järvinen KM. Human milk antibodies to global pathogens reveal geographic and interindividual variations in IgA and IgG. J Clin Invest 2024; 134:e168789. [PMID: 39087469 PMCID: PMC11290967 DOI: 10.1172/jci168789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUNDThe use of high-throughput technologies has enabled rapid advancement in the knowledge of host immune responses to pathogens. Our objective was to compare the repertoire, protection, and maternal factors associated with human milk antibodies to infectious pathogens in different economic and geographic locations.METHODSUsing multipathogen protein microarrays, 878 milk and 94 paired serum samples collected from 695 women in 5 high and low-to-middle income countries (Bangladesh, Finland, Peru, Pakistan, and the United States) were assessed for specific IgA and IgG antibodies to 1,607 proteins from 30 enteric, respiratory, and bloodborne pathogens.RESULTSThe antibody coverage across enteric and respiratory pathogens was highest in Bangladeshi and Pakistani cohorts and lowest in the U.S. and Finland. While some pathogens induced a dominant IgA response (Campylobacter, Klebsiella, Acinetobacter, Cryptosporidium, and pertussis), others elicited both IgA and IgG antibodies in milk and serum, possibly related to the invasiveness of the infection (Shigella, enteropathogenic E. coli "EPEC", Streptococcus pneumoniae, Staphylococcus aureus, and Group B Streptococcus). Besides the differences between economic regions and decreases in concentrations over time, human milk IgA and IgG antibody concentrations were lower in mothers with high BMI and higher parity, respectively. In Bangladeshi infants, a higher specific IgA concentration in human milk was associated with delayed time to rotavirus infection, implying protective properties of antirotavirus antibodies, whereas a higher IgA antibody concentration was associated with greater incidence of Campylobacter infection.CONCLUSIONThis comprehensive assessment of human milk antibody profiles may be used to guide the development of passive protection strategies against infant morbidity and mortality.FUNDINGBill and Melinda Gates Foundation grant OPP1172222 (to KMJ); Bill and Melinda Gates Foundation grant OPP1066764 funded the MDIG trial (to DER); University of Rochester CTSI and Environmental Health Sciences Center funded the Rochester Lifestyle study (to RJL); and R01 AI043596 funded PROVIDE (to WAP).
Collapse
Affiliation(s)
| | - Antti E. Seppo
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine, Rochester, New York, USA
| | | | - Jozelyn Pablo
- Antigen Discovery Incorporated, Irvine, California, USA
| | - Chris Hung
- Antigen Discovery Incorporated, Irvine, California, USA
| | - Andy Teng
- Antigen Discovery Incorporated, Irvine, California, USA
| | | | | | - Amit Oberai
- Antigen Discovery Incorporated, Irvine, California, USA
| | - James Miller
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine, Rochester, New York, USA
| | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Pablo Peñataro Yori
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Anna Kaarina Kukkonen
- New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikael Kuitunen
- New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - L. Beryl Guterman
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Shaun K. Morris
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa G. Pell
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Abdullah Al Mahmud
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Girija Ramakrishan
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Eva Heinz
- Departments of Vector Biology and Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Wellcome Sanger Institute, Parasites and Microbes, Cambridge, UK
| | - Beth D. Kirkpatrick
- Vaccine Testing Center and Department of Microbiology and Molecular Genetics, The University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Abu S.G. Faruque
- Emerging Infection and Parasitology Laboratory, Division of Infectious Diseases, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Rashidul Haque
- Emerging Infection and Parasitology Laboratory, Division of Infectious Diseases, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - R. John Looney
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester School of Medicine, Rochester, New York, USA
| | - Margaret N. Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Erkki Savilahti
- New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Saad B. Omer
- Peter O’Donnell Jr. School of Public Health, Dallas, Texas, USA
| | - Daniel E. Roth
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Hospital for Sick Children, Toronto, Ontario, Canada
| | - William A. Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Kirsi M. Järvinen
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine, Rochester, New York, USA
| |
Collapse
|
14
|
Xu J, Duar RM, Quah B, Gong M, Tin F, Chan P, Sim CK, Tan KH, Chong YS, Gluckman PD, Frese SA, Kyle D, Karnani N. Delayed colonization of Bifidobacterium spp. and low prevalence of B. infantis among infants of Asian ancestry born in Singapore: insights from the GUSTO cohort study. Front Pediatr 2024; 12:1421051. [PMID: 38915873 PMCID: PMC11194334 DOI: 10.3389/fped.2024.1421051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Background The loss of ancestral microbes, or the "disappearing microbiota hypothesis" has been proposed to play a critical role in the rise of inflammatory and immune diseases in developed nations. The effect of this loss is most consequential during early-life, as initial colonizers of the newborn gut contribute significantly to the development of the immune system. Methods In this longitudinal study (day 3, week 3, and month 3 post-birth) of infants of Asian ancestry born in Singapore, we studied how generational immigration status and common perinatal factors affect bifidobacteria and Bifidobacterium longum subsp. infantis (B. infantis) colonization. Cohort registry identifier: NCT01174875. Results Our findings show that first-generation migratory status, perinatal antibiotics usage, and cesarean section birth, significantly influenced the abundance and acquisition of bifidobacteria in the infant gut. Most importantly, 95.6% of the infants surveyed in this study had undetectable B. infantis, an early and beneficial colonizer of infant gut due to its ability to metabolize the wide variety of human milk oligosaccharides present in breastmilk and its ability to shape the development of a healthy immune system. A comparative analysis of B. infantis in 12 countries by their GDP per capita showed a remarkably low prevalence of this microbe in advanced economies, especially Singapore. Conclusion This study provides new insights into infant gut microbiota colonization, showing the impact of generational immigration on early-life gut microbiota acquisition. It also warrants the need to closely monitor the declining prevalence of beneficial microbes such as B. infantis in developed nations and its potential link to increasing autoimmune and allergic diseases.
Collapse
Affiliation(s)
- Jia Xu
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | | | - Baoling Quah
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Min Gong
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Felicia Tin
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Penny Chan
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Clinical Data Engagement, Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Choon Kiat Sim
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
| | - Kok Hian Tan
- SingHealth Duke-NUS Institute for Patient Safety and Quality, Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Yap Seng Chong
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynecology and Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D. Gluckman
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Centre for SPDS Centre for Informed Futures, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Steven A. Frese
- Department of Nutrition, University of Nevada, Reno, NV, United States
| | - David Kyle
- Infinant Health, Inc., Davis, CA, United States
| | - Neerja Karnani
- Department of Human Development, Singapore Institute for Clinical Sciences, Agency for Science (SICS), Technology and Research, Singapore (A*STAR), Singapore, Singapore
- Department of Clinical Data Engagement, Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Brownell J, Lee KE, Chasman D, Gangnon R, Bendixsen CG, Barnes K, Grindle K, Pappas T, Bochkov YA, Dresen A, Hou C, Haslam DB, Seroogy CM, Ong IM, Gern JE. Farm animal exposure, respiratory illnesses, and nasal cell gene expression. J Allergy Clin Immunol 2024; 153:1647-1654. [PMID: 38309597 PMCID: PMC11162314 DOI: 10.1016/j.jaci.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Farm exposures in early life reduce the risks for childhood allergic diseases and asthma. There is less information about how farm exposures relate to respiratory illnesses and mucosal immune development. OBJECTIVE We hypothesized that children raised in farm environments have a lower incidence of respiratory illnesses over the first 2 years of life than nonfarm children. We also analyzed whether farm exposures or respiratory illnesses were related to patterns of nasal cell gene expression. METHODS The Wisconsin Infant Study Cohort included farm (n = 156) and nonfarm (n = 155) families with children followed to age 2 years. Parents reported prenatal farm and other environmental exposures. Illness frequency and severity were assessed using illness diaries and periodic surveys. Nasopharyngeal cell gene expression in a subset of 64 children at age 2 years was compared to farm exposure and respiratory illness history. RESULTS Farm versus nonfarm children had nominally lower rates of respiratory illnesses (rate ratio 0.82 [95% CI, 0.69, 0.97]) with a stepwise reduction in illness rates in children exposed to 0, 1, or ≥2 animal species, but these trends were nonsignificant in a multivariable model. Farm exposures and preceding respiratory illnesses were positively related to nasal cell gene signatures for mononuclear cells and innate and antimicrobial responses. CONCLUSIONS Maternal and infant exposure to farms and farm animals was associated with nonsignificant trends for reduced respiratory illnesses. Nasal cell gene expression in a subset of children suggests that farm exposures and respiratory illnesses in early life are associated with distinct patterns of mucosal immune expression.
Collapse
Affiliation(s)
- Joshua Brownell
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis
| | - Kristine E Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wis
| | - Deborah Chasman
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wis; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wis
| | - Ronald Gangnon
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wis
| | - Casper G Bendixsen
- National Farm Medicine Center, Marshfield Clinic Research Institute, Marshfield, Wis
| | - Katherine Barnes
- National Farm Medicine Center, Marshfield Clinic Research Institute, Marshfield, Wis
| | - Kristine Grindle
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis
| | - Tressa Pappas
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis
| | - Yury A Bochkov
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis
| | - Amy Dresen
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis
| | - Christine Hou
- Department of Statistics, University of Wisconsin-Madison, Madison
| | - David B Haslam
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | | | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wis; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wis
| | - James E Gern
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wis.
| |
Collapse
|
16
|
Paciência I, Sharma N, Hugg TT, Rantala AK, Heibati B, Al-Delaimy WK, Jaakkola MS, Jaakkola JJ. The Role of Biodiversity in the Development of Asthma and Allergic Sensitization: A State-of-the-Science Review. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:66001. [PMID: 38935403 PMCID: PMC11218706 DOI: 10.1289/ehp13948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Changes in land use and climate change have been reported to reduce biodiversity of both the environment and human microbiota. These reductions in biodiversity may lead to inadequate and unbalanced stimulation of immunoregulatory circuits and, ultimately, to clinical diseases, such as asthma and allergies. OBJECTIVE We summarized available empirical evidence on the role of inner (gut, skin, and airways) and outer (air, soil, natural waters, plants, and animals) layers of biodiversity in the development of asthma, wheezing, and allergic sensitization. METHODS We conducted a systematic search in SciVerse Scopus, PubMed MEDLINE, and Web of Science up to 5 March 2024 to identify relevant human studies assessing the relationships between inner and outer layers of biodiversity and the risk of asthma, wheezing, or allergic sensitization. The protocol was registered in PROSPERO (CRD42022381725). RESULTS A total of 2,419 studies were screened and, after exclusions and a full-text review of 447 studies, 82 studies were included in the comprehensive, final review. Twenty-nine studies reported a protective effect of outer layer biodiversity in the development of asthma, wheezing, or allergic sensitization. There were also 16 studies suggesting an effect of outer layer biodiversity on increasing asthma, wheezing, or allergic sensitization. However, there was no clear evidence on the role of inner layer biodiversity in the development of asthma, wheezing, and allergic sensitization (13 studies reported a protective effect and 15 reported evidence of an increased risk). CONCLUSIONS Based on the reviewed literature, a future systematic review could focus more specifically on outer layer biodiversity and asthma. It is unlikely that association with inner layer biodiversity would have enough evidence for systematic review. Based on this comprehensive review, there is a need for population-based longitudinal studies to identify critical periods of exposure in the life course into adulthood and to better understand mechanisms linking environmental exposures and changes in microbiome composition, diversity, and/or function to development of asthma and allergic sensitization. https://doi.org/10.1289/EHP13948.
Collapse
Affiliation(s)
- Inês Paciência
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Needhi Sharma
- University of California, San Diego, San Diego, California, USA
| | - Timo T. Hugg
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Aino K. Rantala
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Behzad Heibati
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Maritta S. Jaakkola
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Jouni J.K. Jaakkola
- Center for Environmental and Respiratory Health Research, Population Health, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Finnish Meteorological Institute, Helsinki, Finland
| |
Collapse
|
17
|
Hilliard MA, Sela DA. Transmission and Persistence of Infant Gut-Associated Bifidobacteria. Microorganisms 2024; 12:879. [PMID: 38792709 PMCID: PMC11124121 DOI: 10.3390/microorganisms12050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Bifidobacterium infantis are the primary colonizers of the infant gut, yet scientific research addressing the transmission of the genus Bifidobacterium to infants remains incomplete. This review examines microbial reservoirs of infant-type Bifidobacterium that potentially contribute to infant gut colonization. Accordingly, strain inheritance from mother to infant via the fecal-oral route is likely contingent on the bifidobacterial strain and phenotype, whereas transmission via the vaginal microbiota may be restricted to Bifidobacterium breve. Additional reservoirs include breastmilk, horizontal transfer from the environment, and potentially in utero transfer. Given that diet is a strong predictor of Bifidobacterium colonization in early life and the absence of Bifidobacterium is observed regardless of breastfeeding, it is likely that additional factors are responsible for bifidobacterial colonization early in life.
Collapse
Affiliation(s)
- Margaret A. Hilliard
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
- Department of Nutrition, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology & Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
18
|
Yang L, Li D, Sun S, Liu D, Wang Y, Liu X, Zhou B, Nie W, Li L, Wang Y, Sha S, Li Y, Shen C, Tao J. Dupilumab therapy improves gut microbiome dysbiosis and tryptophan metabolism in Chinese patients with atopic dermatitis. Int Immunopharmacol 2024; 131:111867. [PMID: 38493690 DOI: 10.1016/j.intimp.2024.111867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Dupilumab has demonstrate its potential to orchestrate inflammatory skin microenvironment, enhance skin barrier and shift skin microbiome dysbiosis, collectively contributing to clinical improvement in patients with atopic dermatitis (AD). As the second genome of human body, growing evidence suggests that the gut microbiome might relate to the host response to treatments. Little is known about the association between dupilumab treatment and gut microbiome in AD patients. OBJECTIVE We aimed to characterize the gut microbiome among Chinese subjects with or without AD and determine the potential effect of dupilumab on the gut microbiome. RESULTS The 16 s rRNA gene sequencing was conducted on 48 healthy controls (HC), 44 AD patients and 27 AD patients who received dupilumab for 16 weeks. Prior to treatment, we identified the changed beta-diversity, increased Firmicutes/Bacteroidetes ratio, decreased Bifidobacterium and expanded Faecalibacterium among the AD patients compared to HC. After 16 weeks of dupilumab treatment, gut microbiome dysbiosis of the AD patients improved with reversed beta-diversity, closer bacterial connections, increased colonization of Bifidobacterium, Ruminococcus gnavus, and Coprococcus, which were negatively correlated with disease severity indicators. This shift was largely independent of the degree of clinical improvement. Bacterial function analysis revealed further metabolic alterations following dupilumab treatment, including up-regulated expression of genes involved in the indole pathway of tryptophan metabolism, corroborated by quantitative UHPLC-MS/MS analysis. CONCLUSION Dupilumab treatment tends to help shift the gut microbial dysbiosis in AD patients to a healthier state, along with improved intestinal tryptophan metabolism, suggesting the gut flora and its metabolites may mediate part of the synergistic therapeutic effects on the host.
Collapse
Affiliation(s)
- Liu Yang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Danqi Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Shuomin Sun
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Danping Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaohuan Liu
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha 410007, Hunan, China
| | - Bin Zhou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Wenjia Nie
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Lu Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Yifei Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Shanshan Sha
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Yan Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Chen Shen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China.
| |
Collapse
|
19
|
Gurung M, Schlegel BT, Rajasundaram D, Fox R, Bode L, Yao T, Lindemann SR, LeRoith T, Read QD, Simecka C, Carroll L, Andres A, Yeruva L. Microbiota from human infants consuming secretors or non-secretors mothers' milk impacts the gut and immune system in mice. mSystems 2024; 9:e0029424. [PMID: 38530054 PMCID: PMC11019842 DOI: 10.1128/msystems.00294-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Maternal secretor status is one of the determinants of human milk oligosaccharides (HMOs) composition, which, in turn, influences the gut microbiota composition of infants. To understand if this change in gut microbiota impacts immune cell composition, intestinal morphology, and gene expression, 21-day-old germ-free C57BL/6 mice were transplanted with fecal microbiota from infants whose mothers were either secretors (SMM) or non-secretors (NSM) or from infants consuming dairy-based formula (MFM). For each group, one set of mice was supplemented with HMOs. HMO supplementation did not significantly impact the microbiota diversity; however, SMM mice had a higher abundance of genus Bacteroides, Bifidobacterium, and Blautia, whereas, in the NSM group, there was a higher abundance of Akkermansia, Enterocloster, and Klebsiella. In MFM, gut microbiota was represented mainly by Parabacteroides, Ruminococcaceae_unclassified, and Clostrodium_sensu_stricto. In mesenteric lymph node, Foxp3+ T cells and innate lymphoid cells type 2 were increased in MFM mice supplemented with HMOs, while in the spleen, they were increased in SMM + HMOs mice. Similarly, serum immunoglobulin A was also elevated in MFM + HMOs group. Distinct global gene expression of the gut was observed in each microbiota group, which was enhanced with HMOs supplementation. Overall, our data show that distinct infant gut microbiota due to maternal secretor status or consumption of dairy-based formula and HMO supplementation impacts immune cell composition, antibody response, and intestinal gene expression in a mouse model. IMPORTANCE Early life factors like neonatal diet modulate gut microbiota, which is important for the optimal gut and immune function. One such factor, human milk oligosaccharides (HMOs), the composition of which is determined by maternal secretor status, has a profound effect on infant gut microbiota. However, how the infant gut microbiota composition determined by maternal secretor status or consumption of infant formula devoid of HMOs impacts infant intestinal ammorphology, gene expression, and immune signature is not well explored. This study provides insights into the differential establishment of infant microbiota derived from infants fed by secretor or non-secretor mothers milk or those consuming infant formula and demonstrates that the secretor status of mothers promotes Bifidobacteria and Bacteroides sps. establishment. This study also shows that supplementation of pooled HMOs in mice changed immune cell composition in the spleen and mesenteric lymph nodes and immunoglobulins in circulation. Hence, this study highlights that maternal secretor status has a role in infant gut microbiota composition, and this, in turn, can impact host gut and immune system.
Collapse
Affiliation(s)
- Manoj Gurung
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
| | - Brent Thomas Schlegel
- University of Pittsburgh Medical Center (UPMC), Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhivyaa Rajasundaram
- University of Pittsburgh Medical Center (UPMC), Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Renee Fox
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Tianming Yao
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, Indiana, USA
| | - Stephen R. Lindemann
- Department of Food Science, Whistler Center for Carbohydrate Research, Purdue University, West Lafayette, Indiana, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, Virginia, USA
| | | | - Christy Simecka
- Division of Laboratory Animal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laura Carroll
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Aline Andres
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Laxmi Yeruva
- Microbiome and Metabolism Research Unit (MMRU), USDA-ARS, SEA, Arkansas Children’s Nutrition Center, Little Rock, Arkansas, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
20
|
Ennis D, Shmorak S, Jantscher-Krenn E, Yassour M. Longitudinal quantification of Bifidobacterium longum subsp. infantis reveals late colonization in the infant gut independent of maternal milk HMO composition. Nat Commun 2024; 15:894. [PMID: 38291346 PMCID: PMC10827747 DOI: 10.1038/s41467-024-45209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/15/2024] [Indexed: 02/01/2024] Open
Abstract
Breast milk contains human milk oligosaccharides (HMOs) that cannot be digested by infants, yet nourish their developing gut microbiome. While Bifidobacterium are the best-known utilizers of individual HMOs, a longitudinal study examining the evolving microbial community at high-resolution coupled with mothers' milk HMO composition is lacking. Here, we developed a high-throughput method to quantify Bifidobacterium longum subsp. infantis (BL. infantis), a proficient HMO-utilizer, and applied it to a longitudinal cohort consisting of 21 mother-infant dyads. We observed substantial changes in the infant gut microbiome over the course of several months, while the HMO composition in mothers' milk remained relatively stable. Although Bifidobacterium species significantly influenced sample variation, no specific HMOs correlated with Bifidobacterium species abundance. Surprisingly, we found that BL. infantis colonization began late in the breastfeeding period both in our cohort and in other geographic locations, highlighting the importance of focusing on BL. infantis dynamics in the infant gut.
Collapse
Affiliation(s)
- Dena Ennis
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimrit Shmorak
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Moran Yassour
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
21
|
Meng X, Shu Q. Novel primers to identify a wider diversity of butyrate-producing bacteria. World J Microbiol Biotechnol 2024; 40:76. [PMID: 38252387 DOI: 10.1007/s11274-023-03872-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024]
Abstract
Butyrate-producing bacteria are a functionally important part of the intestinal tract flora, and the resulting butyric acid is essential for maintaining host intestinal health, regulating the immune system, and influencing energy metabolism. However, butyrate-producing bacteria have not been defined as a coherent phylogenetic group. They are primarily identified using primers for key genes in the butyrate-producing pathway, and their use has been limited to the Bacillota and Bacteroidetes phyla. To overcome this limitation, we developed functional gene primers able to identify butyrate-producing bacteria through the butyrate kinase gene, which encodes the enzyme involved in the final step of the butyrate-producing pathway. Genomes extracted from human and rat feces were used to amplify the target genes through PCR. The obtained sequences were analyzed using BLASTX to construct a developmental tree using the MEGA software. The newly designed butyrate kinase gene primers allowed to recognize a wider diversity of butyrate-producing bacteria than that recognized using currently available primers. Specifically, butyrate-producing bacteria from the Synergistota and Spirochaetota phyla were identified for the first time using these primers. Thus, the developed primers provide a more accurate method for researchers and doctors to identify potential butyrate-producing bacteria and deepen our understanding of butyrate-producing bacterial species.
Collapse
Affiliation(s)
- Xianbin Meng
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Qinglong Shu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
22
|
Ding M, Li B, Chen H, Ross RP, Stanton C, Jiang S, Zhao J, Chen W, Yang B. Bifidobacterium longum subsp. infantis regulates Th1/Th2 balance through the JAK-STAT pathway in growing mice. MICROBIOME RESEARCH REPORTS 2024; 3:16. [PMID: 38841405 PMCID: PMC11149089 DOI: 10.20517/mrr.2023.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 06/07/2024]
Abstract
Objectives: Bifidobacterium longum subsp. infantis is a dominant bacterium in infant gut, which plays a critical role in maintaining the health and development of infants. This study investigated the abilities of eight different strains of B. longum subsp. infantis to regulate the T helper (Th)1/Th2 balance. Methods: Eight B. longum subsp. infantis strains, including I2MI (FJSWXI2MIM1), I4MI [FJSWXI4MI (CCFM1270)], I4MNI (FJSWXI4MNIM1), I5TI (FJSWXI5TIM1), I6TI (FJSWXI6TIM1), I8TI [FJSWXI8TI (CCFM1271)], I10TI [FJSWXI10TI (CCFM1272)], and B6MNI [BJSWXB6MNIM1 (CCFM1269)], were gavaged to BALB/C pups in both female (n = 8) and male (n = 8) mice starting from 1 to 3 weeks old (1 × 109 CFU/day/mice). Selected immune cells were assessed by immunofluorescence and flow cytometry. Cytokines and immunoglobulins were determined by ELISA. Bacterial and bifidobacterial communities were determined by 16S rRNA gene sequencing and bifidobacterial groEL sequencing. Results: B. longum subsp. infantis I4MI and I8TI were shown to increase the ration of colonic IgG2a/IgE in male mice (P < 0.05). B6MNI was demonstrated to significantly increase the levels of colonic IFN-γ and IgG2a, as well as the ratio of IgG2a/IgE in female mice (P < 0.05). It was also shown to significantly increase the ratio of colonic IgG2a/IgE (P < 0.05) and reduce the level of colonic IL-4 in male mice (P < 0.05). Furthermore, B6MNI was demonstrated to regulate colonic JAK/STAT pathway in both male and female mice. I4MI, I5TI, and B6MNI were shown to increase the relative abundance of Bifidobacterium and B. longum subsp. infantis in both male and female mice, whereas I8TI was only shown to increase the relative abundance of Bifidobacterium and B. longum subsp. infantis in male mice (P < 0.05). Conclusion: These results indicated supplementation with B. longum subsp. infantis in early infancy may regulate the Th1/Th2 immune balance, which may prevent the development of related diseases.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - R. Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork T12 R229, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork T12 R229, Ireland
- Teagasc Food Research Centre, Moorepark, Co. Cork P61 C996, Ireland
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing 100015, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing 100083, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
23
|
Laursen MF, Roager HM. Human milk oligosaccharides modify the strength of priority effects in the Bifidobacterium community assembly during infancy. THE ISME JOURNAL 2023; 17:2452-2457. [PMID: 37816852 PMCID: PMC10689826 DOI: 10.1038/s41396-023-01525-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Despite the significant role of the gut microbiota in infant health and development, little is known about the ecological processes determining gut microbial community assembly. According to ecology theory, the timing and order of arrival of microbial species into an ecosystem affect microbial community assembly, a phenomenon termed priority effects. Bifidobacterium species are recognized as highly abundant early colonizers of the infant's gut, partly due to their ability to selectively utilize human milk oligosaccharides (HMOs) from breast milk. However, the role of priority effects in Bifidobacterium community assembly remains unclear. Here, we investigated the Bifidobacterium community assembly in the gut of 25 breastfed Danish infants longitudinally sampled throughout the first 6 months of life. Our results showed that the breastfed infants were often initially, but temporarily, dominated by suboptimal HMO-utilizing Bifidobacterium taxa, such as B. longum subsp. longum, before more efficient HMO-utilizers such as B. longum subsp. infantis, replaced the first colonizer as the dominant Bifidobacterium taxon. Subsequently, we validated this observation using gnotobiotic mice sequentially colonized with B. longum subsp. longum and B. longum subsp. infantis or vice versa, with or without supplementation of HMOs in the drinking water. The results showed that in the absence of HMOs, order of arrival determined dominance. Yet, when mice were supplemented with HMOs the strength of priority effects diminished, and B. longum subsp. infantis dominated regardless of colonization order. Our data demonstrate that the arrival order of Bifidobacterium taxa and the deterministic force of breast milk-derived HMOs, dictate Bifidobacterium community assembly in the infant's gut.
Collapse
Affiliation(s)
- Martin F Laursen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Henrik M Roager
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
24
|
Melsaether C, Høtoft D, Wellejus A, Hermes GDA, Damholt A. Seeding the Infant Gut in Early Life-Effects of Maternal and Infant Seeding with Probiotics on Strain Transfer, Microbiota, and Gastrointestinal Symptoms in Healthy Breastfed Infants. Nutrients 2023; 15:4000. [PMID: 37764787 PMCID: PMC10538230 DOI: 10.3390/nu15184000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
We investigated the effects of two dosing regimens of two multi-strain probiotic products on the gut microbiota of breastfed infants, including the transfer of the dosed strains and clinical outcomes. In forty-seven dyads, infants were either exposed through maternal intake (MS) of Lactobacillus acidophilus LA-5, Bifidobacterium animalis subsp. lactis BB-12, Lacticaseibacillus rhamnosus LGG, and Bifidobacterium longum subsp. infantis Bifin02 from gestational week thirty-three until four weeks after birth (n = 24) or dosed directly (IS) with the same strains except for LA-5 starting within 24 h after birth until day 28 (n = 23). Infant stool samples were collected on day 0, 14, 28, and 42 after birth. Gastrointestinal symptoms were assessed by parents using an electronic diary. Microbiota composition was determined using 16S rRNA sequencing, and strain recovery was analyzed by qPCR. Notably, 100% of the IS infants were colonized with Bifin02 after 14 days as opposed to only 25% of the MS infants. Mean stool frequency was significantly lower in IS infants compared to MS infants and IS infants had softer stools on day 14, 28, and 42. A significantly steeper slope of progression of inconsolable crying and fussing was observed in MS infants compared to IS infants. In conclusion, direct infant seeding induced a faster increase in fecal bifidobacteria abundancy and Bifin02 recovery compared to dosed through the maternal intake.
Collapse
Affiliation(s)
- Cathrine Melsaether
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Diana Høtoft
- Department of Gynecology and Obstetrics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark;
| | - Anja Wellejus
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Gerben D. A. Hermes
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| | - Anders Damholt
- Chr. Hansen A/S, Boege Alle 10-12, 2970 Hoersholm, Denmark; (A.W.); (G.D.A.H.); (A.D.)
| |
Collapse
|
25
|
Donovan SM, Aghaeepour N, Andres A, Azad MB, Becker M, Carlson SE, Järvinen KM, Lin W, Lönnerdal B, Slupsky CM, Steiber AL, Raiten DJ. Evidence for human milk as a biological system and recommendations for study design-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 4. Am J Clin Nutr 2023; 117 Suppl 1:S61-S86. [PMID: 37173061 PMCID: PMC10356565 DOI: 10.1016/j.ajcnut.2022.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 05/15/2023] Open
Abstract
Human milk contains all of the essential nutrients required by the infant within a complex matrix that enhances the bioavailability of many of those nutrients. In addition, human milk is a source of bioactive components, living cells and microbes that facilitate the transition to life outside the womb. Our ability to fully appreciate the importance of this matrix relies on the recognition of short- and long-term health benefits and, as highlighted in previous sections of this supplement, its ecology (i.e., interactions among the lactating parent and breastfed infant as well as within the context of the human milk matrix itself). Designing and interpreting studies to address this complexity depends on the availability of new tools and technologies that account for such complexity. Past efforts have often compared human milk to infant formula, which has provided some insight into the bioactivity of human milk, as a whole, or of individual milk components supplemented with formula. However, this experimental approach cannot capture the contributions of the individual components to the human milk ecology, the interaction between these components within the human milk matrix, or the significance of the matrix itself to enhance human milk bioactivity on outcomes of interest. This paper presents approaches to explore human milk as a biological system and the functional implications of that system and its components. Specifically, we discuss study design and data collection considerations and how emerging analytical technologies, bioinformatics, and systems biology approaches could be applied to advance our understanding of this critical aspect of human biology.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, IL, USA.
| | - Nima Aghaeepour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Department of Pediatrics, and Department of Biomedical Data Sciences, School of Medicine, Stanford University, Stanford, CA, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Meghan B Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Department of Pediatrics and Child Health and Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Martin Becker
- Department of Anesthesiology, Pain, and Perioperative Medicine, Department of Pediatrics, and Department of Biomedical Data Sciences, School of Medicine, Stanford University, Stanford, CA, USA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kirsi M Järvinen
- Department of Pediatrics, Division of Allergy and Immunology and Center for Food Allergy, University of Rochester Medical Center, New York, NY, USA
| | - Weili Lin
- Biomedical Research Imaging Center and Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, CA, USA
| | - Carolyn M Slupsky
- Department of Nutrition, University of California, Davis, CA, USA; Department of Food Science and Technology, University of California, Davis, CA, USA
| | | | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The increasing global prevalence of food allergy indicates that environmental exposures are likely contributing to food allergy development. This review summarizes recent studies on how specific factors within the external exposome may impact the development of food allergy. RECENT FINDINGS There is strong evidence that nonoral exposure to food allergens within the living environment is a risk factor for food sensitization and food allergy. The role of air pollution in food allergy development remains unclear, as cohort studies have not found consistent relationships between air pollutant exposure and food sensitization. Early-life microbial exposures linked to a rural lifestyle are likely protective against food allergy development, possibly through alteration of the infant microbiome. In contrast, factors associated with urbanization and decreased exposure to microbes may contribute to food allergy development. Recent studies on the role of residential greenness in food allergy development suggest either no relationship or a possible increased risk for food allergy. SUMMARY The external exposome comprises a number of exposures that can modify food allergy risk. Improved understanding of how complex environmental exposures interact with genetic factors will be necessary for developing effective interventions aimed at preventing food allergy development in children.
Collapse
Affiliation(s)
- Timothy P. Moran
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Breastfeeding enrichment of B. longum subsp. infantis mitigates the effect of antibiotics on the microbiota and childhood asthma risk. MED 2023; 4:92-112.e5. [PMID: 36603585 DOI: 10.1016/j.medj.2022.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/09/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Early antibiotic exposure is linked to persistent disruption of the infant gut microbiome and subsequent elevated pediatric asthma risk. Breastfeeding acts as a primary modulator of the gut microbiome during early life, but its effect on asthma development has remained unclear. METHODS We harnessed the CHILD cohort to interrogate the influence of breastfeeding on antibiotic-associated asthma risk in a subset of children (n = 2,521). We then profiled the infant microbiomes in a subset of these children (n = 1,338) using shotgun metagenomic sequencing and compared human milk oligosaccharide and fatty acid composition from paired maternal human milk samples for 561 of these infants. FINDINGS Children who took antibiotics without breastfeeding had 3-fold higher asthma odds, whereas there was no such association in children who received antibiotics while breastfeeding. This benefit was associated with widespread "re-balancing" of taxonomic and functional components of the infant microbiome. Functional changes associated with asthma protection were linked to enriched Bifidobacterium longum subsp. infantis colonization. Network analysis identified a selection of fucosylated human milk oligosaccharides in paired maternal samples that were positively associated with B. infantis and these broader functional changes. CONCLUSIONS Our data suggest that breastfeeding and antibiotics have opposing effects on the infant microbiome and that breastfeeding enrichment of B. infantis is associated with reduced antibiotic-associated asthma risk. FUNDING This work was supported in part by the Canadian Institutes of Health Research; the Allergy, Genes and Environment Network of Centres of Excellence; Genome Canada; and Genome British Columbia.
Collapse
|
28
|
Jackson CM, Kaplan AN, Järvinen KM. Environmental Exposures may Hold the Key; Impact of Air Pollution, Greenness, and Rural/Farm Lifestyle on Allergic Outcomes. Curr Allergy Asthma Rep 2023; 23:77-91. [PMID: 36609951 PMCID: PMC9932951 DOI: 10.1007/s11882-022-01061-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW There has been an increased prevalence of allergy. Due to this relatively rapid rise, changes in environmental exposures are likely the main contributor. In this review, we highlight literature from the last 3 years pertaining to the role of air pollution, greenness, and the rural/farm lifestyle and their association with the development of allergic sensitization, atopic dermatitis, food allergy, and allergic rhinitis in infancy and childhood. Because asthma has a more complex pathophysiology, it was excluded from this review. RECENT FINDINGS Recent studies support a role for air pollution, greenness, and rural/farming lifestyle influencing atopic outcomes that continue to be defined. While many studies have examined singular environmental exposures, the interconnectedness of these exposures and others points to a need for future work to consider an individual's whole exposure. Environmental exposures' influence on atopic disease development remains an ongoing and important area of study.
Collapse
Affiliation(s)
- Courtney M Jackson
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Golisano Children's Hospital, 601 Elmwood Ave. Box 777, Rochester, NY, 14642, USA
| | - Alexandra N Kaplan
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Golisano Children's Hospital, 601 Elmwood Ave. Box 777, Rochester, NY, 14642, USA
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Golisano Children's Hospital, 601 Elmwood Ave. Box 777, Rochester, NY, 14642, USA.
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave. Box 777, Rochester, NY, 14642, USA.
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave. Box 777, Rochester, NY, 14642, USA.
| |
Collapse
|
29
|
Derrien M, Mikulic N, Uyoga MA, Chenoll E, Climent E, Howard-Varona A, Nyilima S, Stoffel NU, Karanja S, Kottler R, Stahl B, Zimmermann MB, Bourdet-Sicard R. Gut microbiome function and composition in infants from rural Kenya and association with human milk oligosaccharides. Gut Microbes 2023; 15:2178793. [PMID: 36794816 PMCID: PMC9980514 DOI: 10.1080/19490976.2023.2178793] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
The gut microbiota evolves rapidly after birth, responding dynamically to environmental factors and playing a key role in short- and long-term health. Lifestyle and rurality have been shown to contribute to differences in the gut microbiome, including Bifidobacterium levels, between infants. We studied the composition, function and variability of the gut microbiomes of 6- to 11-month-old Kenyan infants (n = 105). Shotgun metagenomics showed Bifidobacterium longum to be the dominant species. A pangenomic analysis of B. longum in gut metagenomes revealed a high prevalence of B. longum subsp. infantis (B. infantis) in Kenyan infants (80%), and possible co-existence of this subspecies with B. longum subsp. longum. Stratification of the gut microbiome into community (GMC) types revealed differences in composition and functional features. GMC types with a higher prevalence of B. infantis and abundance of B. breve also had a lower pH and a lower abundance of genes encoding pathogenic features. An analysis of human milk oligosaccharides (HMOs) classified the human milk (HM) samples into four groups defined on the basis of secretor and Lewis polymorphisms revealed a higher prevalence of HM group III (Se+, Le-) (22%) than in most previously studied populations, with an enrichment in 2'-fucosyllactose. Our results show that the gut microbiome of partially breastfed Kenyan infants over the age of six months is enriched in bacteria from the Bifidobacterium community, including B. infantis, and that the high prevalence of a specific HM group may indicate a specific HMO-gut microbiome association. This study sheds light on gut microbiome variation in an understudied population with limited exposure to modern microbiome-altering factors.
Collapse
Affiliation(s)
- Muriel Derrien
- Advanced Health & Science, Danone Nutricia Research, Palaiseau, France,CONTACT Muriel Derrien Advanced Health & Science, Danone Nutricia Research, Palaiseau, France
| | - Nadja Mikulic
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Mary A Uyoga
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Empar Chenoll
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Eric Climent
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Adrian Howard-Varona
- ADM-Biopolis, ADM, Parc Cientific Universitat de Valencia, Paterna, Valencia, Spain
| | - Suzane Nyilima
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Simon Karanja
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | | | - Bernd Stahl
- Advanced Health & Science, Danone Nutricia Research, Utrecht, The Netherlands,Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Raphaëlle Bourdet-Sicard
- Advanced Health & Science, Danone Nutricia Research, Palaiseau, France,Raphaëlle Bourdet-Sicard Advanced Health & Science, Danone Nutricia Research, Palaiseau, France
| |
Collapse
|
30
|
Sankova MV, Nikolenko VN, Sankov SV, Sinelnikov MY. SARS-CoV-2 and microbiome. AUTOIMMUNITY, COVID-19, POST-COVID19 SYNDROME AND COVID-19 VACCINATION 2023:279-337. [DOI: 10.1016/b978-0-443-18566-3.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
31
|
Mills DA, German JB, Lebrilla CB, Underwood MA. Translating neonatal microbiome science into commercial innovation: metabolism of human milk oligosaccharides as a basis for probiotic efficacy in breast-fed infants. Gut Microbes 2023; 15:2192458. [PMID: 37013357 PMCID: PMC10075334 DOI: 10.1080/19490976.2023.2192458] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
For over a century, physicians have witnessed a common enrichment of bifidobacteria in the feces of breast-fed infants that was readily associated with infant health status. Recent advances in bacterial genomics, metagenomics, and glycomics have helped explain the nature of this unique enrichment and enabled the tailored use of probiotic supplementation to restore missing bifidobacterial functions in at-risk infants. This review documents a 20-year span of discoveries that set the stage for the current use of human milk oligosaccharide-consuming bifidobacteria to beneficially colonize, modulate, and protect the intestines of at-risk, human milk-fed, neonates. This review also presents a model for probiotic applications wherein bifidobacterial functions, in the form of colonization and HMO-related catabolic activity in situ, represent measurable metabolic outcomes by which probiotic efficacy can be scored toward improving infant health.
Collapse
Affiliation(s)
- David A. Mills
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
- Department of Viticulture and Enology, University of California-Davis, Davis, CA, United States
- Foods for Health Institute, University of California-Davis, Davis, CA, United States
| | - J. Bruce German
- Department of Food Science and Technology, University of California-Davis, Davis, CA, United States
- Foods for Health Institute, University of California-Davis, Davis, CA, United States
| | - Carlito B. Lebrilla
- Foods for Health Institute, University of California-Davis, Davis, CA, United States
- Department of Chemistry, University of California-Davis, Davis, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Davis, CA, United States
| | - Mark A. Underwood
- Foods for Health Institute, University of California-Davis, Davis, CA, United States
- Division of Neonatology, Department of Pediatrics, University of California-Davis, Sacramento, CA, United States
| |
Collapse
|
32
|
Martino C, Dilmore AH, Burcham ZM, Metcalf JL, Jeste D, Knight R. Microbiota succession throughout life from the cradle to the grave. Nat Rev Microbiol 2022; 20:707-720. [PMID: 35906422 DOI: 10.1038/s41579-022-00768-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 11/08/2022]
Abstract
Associations between age and the human microbiota are robust and reproducible. The microbial composition at several body sites can predict human chronological age relatively accurately. Although it is largely unknown why specific microorganisms are more abundant at certain ages, human microbiota research has elucidated a series of microbial community transformations that occur between birth and death. In this Review, we explore microbial succession in the healthy human microbiota from the cradle to the grave. We discuss the stages from primary succession at birth, to disruptions by disease or antibiotic use, to microbial expansion at death. We address how these successions differ by body site and by domain (bacteria, fungi or viruses). We also review experimental tools that microbiota researchers use to conduct this work. Finally, we discuss future directions for studying the microbiota's relationship with age, including designing consistent, well-powered, longitudinal studies, performing robust statistical analyses and improving characterization of non-bacterial microorganisms.
Collapse
Affiliation(s)
- Cameron Martino
- Department of Paediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
| | - Amanda Hazel Dilmore
- Department of Paediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA
- Biomedical Sciences Program, University of California, San Diego, La Jolla, CA, USA
| | - Zachary M Burcham
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jessica L Metcalf
- Department of Animal Sciences, Colorado State University, Fort Collins, CO, USA
| | - Dilip Jeste
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Sam and Rose Stein Institute for Research on Aging, University of California, San Diego, La Jolla, CA, USA
| | - Rob Knight
- Department of Paediatrics, University of California San Diego School of Medicine, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
33
|
Rey-Mariño A, Francino MP. Nutrition, Gut Microbiota, and Allergy Development in Infants. Nutrients 2022; 14:nu14204316. [PMID: 36297000 PMCID: PMC9609088 DOI: 10.3390/nu14204316] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
The process of gut microbiota development in infants is currently being challenged by numerous factors associated with the contemporary lifestyle, including diet. A thorough understanding of all aspects of microbiota development will be necessary for engineering strategies that can modulate it in a beneficial direction. The long-term consequences for human development and health of alterations in the succession pattern that forms the gut microbiota are just beginning to be explored and require much further investigation. Nevertheless, it is clear that gut microbiota development in infancy bears strong associations with the risk for allergic disease. A useful understanding of microbial succession in the gut of infants needs to reveal not only changes in taxonomic composition but also the development of functional capacities through time and how these are related to diet and various environmental factors. Metagenomic and metatranscriptomic studies have started to produce insights into the trends of functional repertoire and gene expression change within the first year after birth. This understanding is critical as during this period the most substantial development of the gut microbiota takes place and the relations between gut microbes and host immunity are established. However, further research needs to focus on the impact of diet on these changes and on how diet can be used to counteract the challenges posed by modern lifestyles to microbiota development and reduce the risk of allergic disease.
Collapse
Affiliation(s)
- Alejandra Rey-Mariño
- Genomics and Health Department, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region (FISABIO), 46020 València, Spain
| | - M. Pilar Francino
- Genomics and Health Department, Foundation for the Promotion of Health and Biomedical Research of the Valencia Region (FISABIO), 46020 València, Spain
- CIBER en Epidemiología y Salud Pública (CIBERESP), 28001 Madrid, Spain
- Correspondence:
| |
Collapse
|
34
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
35
|
Davis EC, Jackson CM, Ting T, Harizaj A, Järvinen KM. Predictors and biomarkers of food allergy and sensitization in early childhood. Ann Allergy Asthma Immunol 2022; 129:292-300. [PMID: 35490857 PMCID: PMC11910167 DOI: 10.1016/j.anai.2022.04.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To review existing literature on the early risk factors for and biomarkers of food allergy (FA) and food sensitization (FS) and highlight opportunities for future research that will further the understanding of FA pathogenesis in infancy and toddlerhood. DATA SOURCES PubMed search of English-language articles related to FA and atopic disease. STUDY SELECTIONS Human studies with outcomes related to FA, FS, and other atopic disease in childhood were selected and reviewed. Studies published after 2015 were prioritized. RESULTS The prevalence of FA has greatly increased in recent decades and is now a global public health concern. A complex network of early life risk factors has been associated with development of FA and FS in childhood. Food allergy has a genetic component, but recent evidence suggests that interactions between risk alleles and other environmental exposures are important for disease pathogenesis, potentially through epigenetic mechanisms. Lifestyle factors, such as delivery mode, antibiotic use, and pet exposure also influence FA risk, which may be through their effect on the early life gut microbiome. How these early life risk factors, along with route and timing of antigen exposure, collectively target the developing immune system remains an ongoing and important area of study. CONCLUSION The current body of evidence emphasizes the first 1000 days of life as a critical period for FA development. More observational studies and adequately powered clinical trials spanning early pregnancy through childhood are needed to identify novel biomarkers and risk factors that can predict susceptibility toward or protection against FA.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Courtney M Jackson
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Tiffany Ting
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York
| | - Albana Harizaj
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York.
| |
Collapse
|
36
|
Järvinen KM, Davis EC, Bevec E, Jackson CM, Pizzarello C, Catlin E, Klein M, Sunkara A, Diaz N, Miller J, Martina CA, Thakar J, Seppo AE, Looney RJ. Biomarkers of Development of Immunity and Allergic Diseases in Farming and Non-farming Lifestyle Infants: Design, Methods and 1 Year Outcomes in the "Zooming in to Old Order Mennonites" Birth Cohort Study. Front Pediatr 2022; 10:916184. [PMID: 35874571 PMCID: PMC9299374 DOI: 10.3389/fped.2022.916184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Traditional farming lifestyle has been shown to be protective against asthma and allergic diseases. The individual factors that appear to be associated with this "farm-life effect" include consumption of unpasteurized farm milk and exposure to farm animals and stables. However, the biomarkers of the protective immunity and those associated with early development of allergic diseases in infancy remain unclear. The "Zooming in to Old Order Mennonites (ZOOM)" study was designed to assess the differences in the lifestyle and the development of the microbiome, systemic and mucosal immunity between infants born to traditional farming lifestyle at low risk for allergic diseases and those born to urban/suburban atopic families with a high risk for allergic diseases in order to identify biomarkers of development of allergic diseases in infancy. 190 mothers and their infants born to Old Order Mennonite population protected from or in Rochester families at high risk for allergic diseases were recruited before birth from the Finger Lakes Region of New York State. Questionnaires and samples are collected from mothers during pregnancy and after delivery and from infants at birth and at 1-2 weeks, 6 weeks, 6, 12, 18, and 24 months, with 3-, 4-, and 5-year follow-up ongoing. Samples collected include maternal blood, stool, saliva, nasal and skin swabs and urine during pregnancy; breast milk postnatally; infant blood, stool, saliva, nasal and skin swabs. Signs and symptoms of allergic diseases are assessed at every visit and serum specific IgE is measured at 1 and 2 years of age. Allergic diseases are diagnosed by clinical history, exam, and sensitization by skin prick test and/or serum specific IgE. By the end of the first year of life, the prevalence of food allergy and atopic dermatitis were higher in ROC infants compared to the rates observed in OOM infants as was the number of infants sensitized to foods. These studies of immune system development in a population protected from and in those at risk for allergic diseases will provide critical new knowledge about the development of the mucosal and systemic immunity and lay the groundwork for future studies of prevention of allergic diseases.
Collapse
Affiliation(s)
- Kirsi M. Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Erin C. Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Erin Bevec
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Courtney M. Jackson
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Catherine Pizzarello
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Elizabeth Catlin
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Miranda Klein
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Akhila Sunkara
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Nichole Diaz
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - James Miller
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - Camille A. Martina
- Department of Public Health and Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Juilee Thakar
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Antti E. Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, NY, United States
| | - R. John Looney
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
37
|
Holm M, Saraswat M, Joenväärä S, Seppo A, Looney RJ, Tohmola T, Renkonen J, Renkonen R, Järvinen KM. Quantitative glycoproteomics of human milk and association with atopic disease. PLoS One 2022; 17:e0267967. [PMID: 35559953 PMCID: PMC9106177 DOI: 10.1371/journal.pone.0267967] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
The prevalence of allergic diseases and asthma is increasing rapidly worldwide, with environmental and lifestyle behaviors implicated as a reason. Epidemiological studies have shown that children who grow up on farms are at lower risk of developing childhood atopic disease, indicating the presence of a protective "farm effect". The Old Order Mennonite (OOM) community in Upstate New York have traditional, agrarian lifestyles, a low rate of atopic disease, and long periods of exclusive breastfeeding. Human milk proteins are heavily glycosylated, although there is a paucity of studies investigating the milk glycoproteome. In this study, we have used quantitative glycoproteomics to compare the N-glycoprotein profiles of 54 milk samples from Rochester urban/suburban and OOM mothers, two populations with different lifestyles, exposures, and risk of atopic disease. We also compared N-glycoprotein profiles according to the presence or absence of atopic disease in the mothers and, separately, the children. We identified 79 N-glycopeptides from 15 different proteins and found that proteins including immunoglobulin A1, polymeric immunoglobulin receptor, and lactotransferrin displayed significant glycan heterogeneity. We found that the abundances of 38 glycopeptides differed significantly between Rochester and OOM mothers and also identified four glycopeptides with significantly different abundances between all comparisons. These four glycopeptides may be associated with the development of atopic disease. The findings of this study suggest that the differential glycosylation of milk proteins could be linked to atopic disease.
Collapse
Affiliation(s)
- Matilda Holm
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mayank Saraswat
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sakari Joenväärä
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Seppo
- Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, New York, United States of America
| | - R. John Looney
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Tiialotta Tohmola
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jutta Renkonen
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Risto Renkonen
- Transplantation Laboratory, Haartman Institute, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kirsi M. Järvinen
- Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Golisano Children’s Hospital, Rochester, New York, United States of America
| |
Collapse
|
38
|
Intestinal ‘Infant-Type’ Bifidobacteria Mediate Immune System Development in the First 1000 Days of Life. Nutrients 2022; 14:nu14071498. [PMID: 35406110 PMCID: PMC9002861 DOI: 10.3390/nu14071498] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 01/05/2023] Open
Abstract
Immune system maturation begins early in life, but few studies have examined how early-life gut microbiota colonization educates the neonatal immune system. Bifidobacteria predominate in the intestines of breastfed infants and metabolize human milk oligosaccharides. This glycolytic activity alters the intestinal microenvironment and consequently stimulates immune system maturation at the neonatal stage. However, few studies have provided mechanistic insights into the contribution of ‘infant-type’ Bifidobacterium species, especially via metabolites such as short-chain fatty acids. In this review, we highlight the first 1000 days of life, which provide a window of opportunity for infant-type bifidobacteria to educate the neonatal immune system. Furthermore, we discuss the instrumental role of infant-type bifidobacteria in the education of the neonatal immune system by inducing immune tolerance and suppressing intestinal inflammation, and the potential underlying mechanism of this immune effect in the first 1000 days of life. We also summarize recent research that suggests the administration of infant-type bifidobacteria helps to modify the intestinal microecology and prevent the progress of immune-mediated disorders.
Collapse
|
39
|
Taft DH, Lewis ZT, Nguyen N, Ho S, Masarweh C, Dunne-Castagna V, Tancredi DJ, Huda MN, Stephensen CB, Hinde K, von Mutius E, Kirjavainen PV, Dalphin JC, Lauener R, Riedler J, Smilowitz JT, German JB, Morrow AL, Mills DA. Bifidobacterium Species Colonization in Infancy: A Global Cross-Sectional Comparison by Population History of Breastfeeding. Nutrients 2022; 14:nu14071423. [PMID: 35406036 PMCID: PMC9003546 DOI: 10.3390/nu14071423] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Bifidobacterium species are beneficial and dominant members of the breastfed infant gut microbiome; however, their health benefits are partially species-dependent. Here, we characterize the species and subspecies of Bifidobacterium in breastfed infants around the world to consider the potential impact of a historic dietary shift on the disappearance of B. longum subsp. infantis in some populations. Across populations, three distinct patterns of Bifidobacterium colonization emerged: (1) The dominance of Bifidobacterium longum subspecies infantis, (2) prevalent Bifidobacterium of multiple species, and (3) the frequent absence of any Bifidobacterium. These patterns appear related to a country’s history of breastfeeding, with infants in countries with historically high rates of long-duration breastfeeding more likely to be colonized by B. longum subspecies infantis compared with infants in countries with histories of shorter-duration breastfeeding. In addition, the timing of infant colonization with B. longum subsp. infantis is consistent with horizontal transmission of this subspecies, rather than the vertical transmission previously reported for other Bifidobacterium species. These findings highlight the need to consider historical and cultural influences on the prevalence of gut commensals and the need to understand epidemiological transmission patterns of Bifidobacterium and other major commensals.
Collapse
Affiliation(s)
- Diana H. Taft
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - Zachery T. Lewis
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Nhu Nguyen
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Steve Ho
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Chad Masarweh
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Vanessa Dunne-Castagna
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
| | - Daniel J. Tancredi
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
| | - M. Nazmul Huda
- US Department of Agriculture, Western Human Nutrition Research Center, Davis, CA 95616, USA; (M.N.H.); (C.B.S.)
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Charles B. Stephensen
- US Department of Agriculture, Western Human Nutrition Research Center, Davis, CA 95616, USA; (M.N.H.); (C.B.S.)
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Katie Hinde
- Center for Evolution and Medicine, School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85281, USA;
| | - Erika von Mutius
- Dr. von Hauner Children’s Hospital, Ludwig Maximilian University, 80337 Munich, Germany;
- Institute for Asthma and Allergy Prevention, Helmholtz Centre Munich, 85764 Neuherberg, Germany
| | - Pirkka V. Kirjavainen
- Environment Health Unit, National Institute for Health and Welfare, 70210 Kuopio, Finland;
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jean-Charles Dalphin
- Department of Respiratory Disease, UMR/CNRS 6249 Chrono-Environment, University Hospital of Besançon, F-25000 Besançon, France;
| | - Roger Lauener
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland;
- Children’s Hospital of Eastern Switzerland, 9000 St. Gallen, Switzerland
| | - Josef Riedler
- Children’s Hospital Schwarzach, 5620 Schwarzach, Austria;
| | - Jennifer T. Smilowitz
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - J. Bruce German
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Foods for Health Institute, University of California Davis, Davis, CA 95616, USA
| | - Ardythe L. Morrow
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
- Correspondence: (A.L.M.); (D.A.M.); Tel.: +1-513-558-0809 (A.L.M.); +1-530-754-7821 (D.A.M.)
| | - David A. Mills
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA; (D.H.T.); (Z.T.L.); (N.N.); (S.H.); (C.M.); (V.D.-C.); (J.T.S.); (J.B.G.)
- Department of Viticulture and Enology, University of California Davis, Davis, CA 95616, USA
- Correspondence: (A.L.M.); (D.A.M.); Tel.: +1-513-558-0809 (A.L.M.); +1-530-754-7821 (D.A.M.)
| |
Collapse
|
40
|
Jackson CM, Mahmood MM, Järvinen KM. Farming lifestyle and human milk: Modulation of the infant microbiome and protection against allergy. Acta Paediatr 2022; 111:54-58. [PMID: 34626494 PMCID: PMC8678317 DOI: 10.1111/apa.16147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
There has been an increased prevalence of several allergic manifestations such as food allergy, atopic eczema, allergic rhinitis and asthma. Several explanations have been proposed why this has occurred, but one of the main contributing factors may be the gradual loss of microbial exposures over time in regions where allergy is prevalent. Such exposures occur in individuals who practise a traditional farming lifestyle and are protected against allergy. Infant consumption of human milk, more commonly practised in these farming communities, may provide an alternative in combatting allergy, as it known to be beneficial to infant health. In this review, we cover human milk and its role in shaping the gut microbiome promoting the growth of beneficial bacteria like Bifidobacterium, as well as the downstream impact of the farming lifestyle, human milk and Bifidobacterium has on developing infant immunity.
Collapse
Affiliation(s)
- Courtney M. Jackson
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Mustafa M. Mahmood
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Kirsi M. Järvinen
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| |
Collapse
|
41
|
Nourishing the Human Holobiont to Reduce the Risk of Non-Communicable Diseases: A Cow’s Milk Evidence Map Example. Appl Microbiol 2021. [DOI: 10.3390/applmicrobiol2010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The microbiome revolution brought the realization that diet, health, and safety for humans in reality means diet, health, and safety for the human holobiont/superorganism. Eating healthier means much more than just feeding human cells. Our diet must also nourish the combination of our microbiome and our connected physiological systems (e.g., the microimmunosome). For this reason, there has been an interest in returning to ancestral “complete” unprocessed foods enriched in microbes, including raw milks. To contribute to this inevitable “nourishing the holobiont” trend, we introduce a systematic risk–benefit analysis tool (evidence mapping), which facilitates transdisciplinary state-of-the-science decisions that transcend single scientific disciplines. Our prior paper developed an evidence map (a type of risk–benefit mind map) for raw vs. processed/pasteurized human breast milk. In the present paper, we follow with a comprehensive evidence map and narrative for raw/natural vs. processed/pasteurized cow’s milk. Importantly, the evidence maps incorporate clinical data for both infectious and non-communicable diseases and allow the impact of modern agricultural, food management, and medical and veterinary monitoring outcomes to be captured. Additionally, we focus on the impact of raw milks (as “complete” foods) on the microimmunosome, the microbiome-systems biology unit that significantly determines risk of the world’s number one cause of human death, non-communicable diseases.
Collapse
|
42
|
Seppo AE, Choudhury R, Pizzarello C, Palli R, Fridy S, Rajani PS, Stern J, Martina C, Yonemitsu C, Bode L, Bu K, Tamburini S, Piras E, Wallach DS, Allen M, Looney RJ, Clemente JC, Thakar J, Järvinen KM. Traditional Farming Lifestyle in Old Older Mennonites Modulates Human Milk Composition. Front Immunol 2021; 12:741513. [PMID: 34707611 PMCID: PMC8545059 DOI: 10.3389/fimmu.2021.741513] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/01/2021] [Indexed: 01/11/2023] Open
Abstract
Background In addition to farming exposures in childhood, maternal farming exposures provide strong protection against allergic disease in their children; however, the effect of farming lifestyle on human milk (HM) composition is unknown. Objective This study aims to characterize the maternal immune effects of Old Order Mennonite (OOM) traditional farming lifestyle when compared with Rochester (ROC) families at higher risk for asthma and allergic diseases using HM as a proxy. Methods HM samples collected at median 2 months of lactation from 52 OOM and 29 ROC mothers were assayed for IgA1 and IgA2 antibodies, cytokines, endotoxin, HM oligosaccharides (HMOs), and targeted fatty acid (FA) metabolites. Development of early childhood atopic diseases in children by 3 years of age was assessed. In addition to group comparisons, systems level network analysis was performed to identify communities of multiple HM factors in ROC and OOM lifestyle. Results HM contains IgA1 and IgA2 antibodies broadly recognizing food, inhalant, and bacterial antigens. OOM HM has significantly higher levels of IgA to peanut, ovalbumin, dust mites, and Streptococcus equii as well TGF-β2, and IFN-λ3. A strong correlation occurred between maternal antibiotic use and levels of several HMOs. Path-based analysis of HMOs shows lower activity in the path involving lactoneohexaose (LNH) in the OOM as well as higher levels of lacto-N-neotetraose (LNnT) and two long-chain FAs C-18OH (stearic acid) and C-23OH (tricosanoic acid) compared with Rochester HM. OOM and Rochester milk formed five different clusters, e.g., butyrate production was associated with Prevotellaceae, Veillonellaceae, and Micrococcaceae cluster. Development of atopic disease in early childhood was more common in Rochester and associated with lower levels of total IgA, IgA2 to dust mite, as well as of TSLP. Conclusion Traditional, agrarian lifestyle, and antibiotic use are strong regulators of maternally derived immune and metabolic factors, which may have downstream implications for postnatal developmental programming of infant's gut microbiome and immune system.
Collapse
Affiliation(s)
- Antti E. Seppo
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States
| | - Rakin Choudhury
- Department of Microbiology and Immunology and Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Catherine Pizzarello
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Sade Fridy
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States
| | - Puja Sood Rajani
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States
| | - Jessica Stern
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States
| | - Camille Martina
- Department of Public Health Sciences & Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Chloe Yonemitsu
- Division of Neonatology and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Lars Bode
- Division of Neonatology and Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States,Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, United States
| | - Kevin Bu
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Precision Immunology Institue, Icahn School of Medicine at Mount Sinai, New York, New York, NY, United States
| | - Sabrina Tamburini
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Precision Immunology Institue, Icahn School of Medicine at Mount Sinai, New York, New York, NY, United States
| | - Enrica Piras
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Precision Immunology Institue, Icahn School of Medicine at Mount Sinai, New York, New York, NY, United States
| | - David S. Wallach
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Precision Immunology Institue, Icahn School of Medicine at Mount Sinai, New York, New York, NY, United States
| | - Maria Allen
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - R. John Looney
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genomic Technology, Precision Immunology Institue, Icahn School of Medicine at Mount Sinai, New York, New York, NY, United States
| | - Juilee Thakar
- Department of Microbiology and Immunology and Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Kirsi M. Järvinen
- Division of Allergy and Immunology and Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry and Golisano Children’s Hospital, Rochester, NY, United States,Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States,*Correspondence: Kirsi M. Järvinen,
| |
Collapse
|
43
|
Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Arzoomand A, Olin A, Wang J, Mikes J, Tan Z, Chen Y, Ehrlich AM, Bernhardsson AK, Mugabo CH, Ambrosiani Y, Gustafsson A, Chew S, Brown HK, Prambs J, Bohlin K, Mitchell RD, Underwood MA, Smilowitz JT, German JB, Frese SA, Brodin P. Bifidobacteria-mediated immune system imprinting early in life. Cell 2021; 184:3884-3898.e11. [PMID: 34143954 DOI: 10.1016/j.cell.2021.05.030] [Citation(s) in RCA: 402] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/19/2021] [Accepted: 05/19/2021] [Indexed: 02/08/2023]
Abstract
Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon β (IFNβ) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.
Collapse
Affiliation(s)
- Bethany M Henrick
- Evolve BioSystems, Inc., Davis, CA 95618, USA; Department of Food Science and Technology, University of Nebraska, Lincoln, Lincoln, NE 68588-6205, USA.
| | - Lucie Rodriguez
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Tadepally Lakshmikanth
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Christian Pou
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ewa Henckel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Aron Arzoomand
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Axel Olin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Jun Wang
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Jaromir Mikes
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ziyang Tan
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Yang Chen
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | | | - Anna Karin Bernhardsson
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Constantin Habimana Mugabo
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden
| | - Ylva Ambrosiani
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Anna Gustafsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | | | | | | | - Kajsa Bohlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14152 Stockholm, Sweden; Department of Neonatology, Karolinska University Hospital, 14186 Stockholm, Sweden
| | | | - Mark A Underwood
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Pediatrics, University of California Davis Children's Hospital, Sacramento, CA 95817, USA
| | - Jennifer T Smilowitz
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, Davis, CA 95616, USA
| | - J Bruce German
- Foods for Health Institute, University of California, Davis, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, Davis, CA 95616, USA
| | - Steven A Frese
- Department of Food Science and Technology, University of Nebraska, Lincoln, Lincoln, NE 68588-6205, USA; Department of Nutrition, University of Nevada, Reno, Reno, NV 89557, USA
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden; Pediatric Rheumatology, Karolinska University Hospital, 17176 Solna, Sweden.
| |
Collapse
|