1
|
Peer A, Samuelson DR. The Role of the Microbiome in Allergy, Asthma, and Occupational Lung Disease. Curr Allergy Asthma Rep 2024; 24:415-423. [PMID: 38904934 PMCID: PMC11297072 DOI: 10.1007/s11882-024-01156-8] [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] [Accepted: 06/07/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW The human commensal microbiota is now widely accepted as a key regulator of human health and disease. The composition of the mucosal associated microbiota has been shown to play a critical role in the lung health. The role of the mucosal microbiota in the development and severity of allergy, asthma, and occupational lung disease is only beginning to take shape. However, advances in our understanding of these links have tremendous potential to led to new clinical interventions to reduce allergy, asthma, and occupational lung disease morbidity. RECENT FINDINGS We review recent work describing the relationship and role of the commensal microbiota in the development of allergy, asthma, and occupational lung disease. Our review primarily focuses on occupational exposures and the effects of the microbiome, both in composition and function. Data generated from these studies may lead to the development of interventions targeted at establishing and maintaining a healthy microbiota. We also highlight the role of environmental exposures and the effects on the commensal microbial community and their potential association with occupational lung disease. This review explores the current research describing the role of the human microbiome in the regulation of pulmonary health and disease, with a specific focus on the role of the mucosal microbiota in the development of allergy, asthma, and occupational lung disease.
Collapse
Affiliation(s)
- Ashley Peer
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep, University of Nebraska Medical Center, Omaha, NE, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep, University of Nebraska Medical Center, Omaha, NE, USA.
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
2
|
Burnham CM, McKenney EA, van Heugten KA, Minter LJ, Trivedi S. Effects of age, seasonality, and reproductive status on the gut microbiome of Southern White Rhinoceros (Ceratotherium simum simum) at the North Carolina zoo. Anim Microbiome 2023; 5:27. [PMID: 37147724 PMCID: PMC10163733 DOI: 10.1186/s42523-023-00249-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 04/22/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Managed southern white rhinoceros (Ceratotherium simum simum) serve as assurance populations for wild conspecifics threatened by poaching and other anthropocentric effects, though many managed populations experience subfertility and reproductive failure. Gut microbiome and host health are inextricably linked, and reproductive outcomes in managed southern white rhinoceros may be mediated in part by their diet and gut microbial diversity. Thus, understanding microbial dynamics within managed populations may help improve conservation efforts. We characterized the taxonomic composition of the gut microbiome in the managed population of female southern white rhinoceros (n = 8) at the North Carolina Zoo and investigated the effects of seasonality (summer vs. winter) and age classes (juveniles (n = 2; 0-2 years), subadults (n = 2; 3-7 years), and adults (n = 4; >7 years)) on microbial richness and community structure. Collection of a fecal sample was attempted for each individual once per month from July-September 2020 and January-March 2021 resulting in a total of 41 samples analyzed. Microbial DNA was extracted and sequenced using the V3-V4 region of the 16S rRNA bacterial gene. Total operational taxonomic units (OTUs), alpha diversity (species richness, Shannon diversity), and beta diversity (Bray-Curtis dissimilarity, linear discriminant analysis effect size) indices were examined, and differentially enriched taxa were identified. RESULTS There were differences (p < 0.05) in alpha and beta diversity indices across individuals, age groups, and sampling months. Subadult females had higher levels of Shannon diversity (Wilcoxon, p < 0.05) compared to adult females and harbored a community cluster distinct from both juveniles and adults. Samples collected during winter months (January-March 2021) possessed higher species richness and statistically distinct communities compared to summer months (July-September 2020) (PERMANOVA, p < 0.05). Reproductively active (n = 2) and currently nonreproductive adult females (n = 2) harbored differentially enriched taxa, with the gut microbiome of nonreproductive females significantly enriched (p = 0.001) in unclassified members of Mobiluncus, a genus which possesses species associated with poor reproductive outcomes in other animal species when identified in the cervicovaginal microbiome. CONCLUSION Together, our results increase the understanding of age and season related microbial variation in southern white rhinoceros at the North Carolina Zoo and have identified a potential microbial biomarker for reproductive concern within managed female southern white rhinoceros.
Collapse
Affiliation(s)
- Christina M Burnham
- Department of Animal Science, North Carolina State University, 120 W Broughton Dr, Raleigh, NC, 27607, USA
| | - Erin A McKenney
- Department of Applied Ecology, North Carolina State University, 100 Brooks Ave, Raleigh, NC, 27607, USA
| | - Kimberly Ange- van Heugten
- Department of Animal Science, North Carolina State University, 120 W Broughton Dr, Raleigh, NC, 27607, USA
| | - Larry J Minter
- North Carolina Zoo, 4401 Zoo Parkway, Asheboro, NC, 27205, USA
| | - Shweta Trivedi
- Department of Animal Science, North Carolina State University, 120 W Broughton Dr, Raleigh, NC, 27607, USA.
| |
Collapse
|
3
|
Desai D, Desai A, Jamil A, Csendes D, Gutlapalli SD, Prakash K, Swarnakari KM, Bai M, Manoharan MP, Raja R, Khan S. Re-defining the Gut Heart Axis: A Systematic Review of the Literature on the Role of Gut Microbial Dysbiosis in Patients With Heart Failure. Cureus 2023; 15:e34902. [PMID: 36938237 PMCID: PMC10014482 DOI: 10.7759/cureus.34902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/10/2023] [Indexed: 02/14/2023] Open
Abstract
Heart failure (HF) contributes to the cardiovascular health burden worldwide. Patients with heart failure have been recently studied to possess unique changes in the gut microbiome that affect immune homeostasis and metabolism. In this systematic review of the literature, we aim to identify the impact of gut dysbiosis on heart failure. We used Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines to conduct our systematic review. We searched the literature on databases such as PubMed, PubMed Central (PMC), Medline, and ScienceDirect. Ten articles were included for review. There were significant differences in the gut microbiome composition in heart failure. Relative abundance of Ruminococcus gnavus, Escherichia Shigella, Streptococcus sp, Veillonella sp, and Actinobacteria, and relative depletion of Eubacterium, Prevotella, Faecalibacterium, SMB53, and Megamonas. The composition varied according to age, heart failure stage, and decompensation level. The composition remained unaltered with ejection fraction. There was an increased expression of genes responsible for the metabolism of amino acids, carbohydrates, choline trimethylamine-lyase (TMA-lyase), lipopolysaccharide (LPS) biosynthesis, tryptophan, and lipid metabolism. The resultant changes affected the levels of metabolites, such as trimethylamine N-oxide (TMAO), indoxyl sulfate (IS), and LPS, and inflammatory markers in the feces and plasma, which contributed to heart failure. These biomarkers of heart failure could serve as targets for the prevention and treatment of heart failure. Patients with heart failure harbor a unique constellation of gut microbiota that affect the pathogenesis of heart failure. Further studies are needed to understand the causal relationship between dysbiosis and heart failure.
Collapse
Affiliation(s)
- Darshi Desai
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, University of California Riverside School of Medicine, St. Bernardine's Medical Center, San Bernardino, USA
| | - Aditya Desai
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, University of California Riverside School of Medicine, St. Bernardine's Medical Center, San Bernardino, USA
| | - Aneeque Jamil
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Denise Csendes
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sai D Gutlapalli
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Keerthana Prakash
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kiran M Swarnakari
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Meena Bai
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mohana P Manoharan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rabab Raja
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Medicine, All Saints University School of Medicine, Roseau, DMA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
4
|
Gildner TE, Cepon-Robins TJ. Rural Embodiment and Community Health: an Anthropological Case Study on Biocultural Determinants of Tropical Disease Infection and Immune System Development in the USA. CURRENT TROPICAL MEDICINE REPORTS 2023; 10:26-39. [PMID: 36714157 PMCID: PMC9868515 DOI: 10.1007/s40475-023-00282-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/24/2023]
Abstract
Purpose of Review Biocultural methods are critically important for identifying environmental and socioeconomic factors linked with tropical disease risk and outcomes. For example, embodiment theory refers to the process by which lived experiences impact individual biology. Increased exposure to pathogens, chronic psychosocial stress, and unequal resource access are all outcomes linked with discrimination and poverty. Through lived experiences, race and socioeconomic inequality can literally become embodied-get under the skin and affect physiology-impacting immune responses and contributing to lifelong health disparities. Yet, few studies have investigated tropical disease patterns and associated immune function using embodiment theory to understand lasting physiological impacts associated with living in a high-pathogen environment. Recent Findings Here, we use preliminary data drawn from the Rural Embodiment and Community Health (REACH) study to assess whether pathogen exposure and immune stimulation within a sample of children from the Mississippi Delta are associated with household income. We also test whether immune marker levels-assessed with enzyme-linked immunosorbent assays using dried blood spot samples-vary between the REACH sample and a similarly aged nationally representative NHANES sample. Immune marker levels did not differ significantly between REACH participants living below vs. above the federal poverty line, yet immunoglobulin E levels-a marker of macroparasite infection-were higher among REACH study participants compared to the NHANES sample. Summary These results may suggest community-level pathogenic exposures (i.e., parasitic infections) are embodied by REACH participants with implications for long-term immune function, potentially resulting in immune aspects that differ from nationally representative samples. Supplementary Information The online version contains supplementary material available at 10.1007/s40475-023-00282-z.
Collapse
Affiliation(s)
- Theresa E Gildner
- Department of Anthropology, Washington University in St. Louis, St. Louis, MO USA
| | - Tara J Cepon-Robins
- Department of Anthropology, University of Colorado Colorado Springs, Colorado Springs, CO USA
| |
Collapse
|
5
|
Naik SS, Ramphall S, Rijal S, Prakash V, Ekladios H, Mulayamkuzhiyil Saju J, Mandal N, Kham NI, Shahid R, Venugopal S. Association of Gut Microbial Dysbiosis and Hypertension: A Systematic Review. Cureus 2022; 14:e29927. [PMID: 36381851 PMCID: PMC9642844 DOI: 10.7759/cureus.29927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/04/2022] [Indexed: 01/25/2023] Open
Abstract
Hypertension (HTN) is one of the most prevalent and dangerous cardiovascular diseases worldwide. Recently, its direct or indirect association with gut dysbiosis has been an interest of study for many. It also includes the metabolomic and functional gene changes in hypertensives compared with healthy individuals. This systematic review aims to study quantitative and qualitative interactions between the two and re-defining the heart-gut axis. We have strictly followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), 2020, guidelines. We conducted an in-depth search of databases such as PubMed, PubMed Central (PMC), Medline, and ScienceDirect to find relevant studies for our topic of interest. After the final quality check, we included eight articles in the systematic review. A significant difference in richness and diversity in gut microbiota was observed in hypertensive patients compared with healthy controls. There was an increased abundance of many bacteria such as Catabacter, Robinsoleilla, Serratia, Enterobacteriaceae, Ruminococcus torques, Parasutterella, Escherichia, Shigella, and Klebsiella, while a decreased abundance of Sporobacter, Roseburia hominis, Romboutsia spp., and Roseburia. Alteration of the composition also varied based on diet, age, ethnicity, and severity of HTN. Short-chain fatty acids (SCFAs)-producing bacteria are found to be on the lower side in hypertensives owing to the protective property of SCFAs against inflammation, especially butyric acid. From the perspective of metabolomic changes, harmful metabolites for cardiovascular health such as intestinal fatty acid binding protein (I-FABP), lipopolysaccharides (LPSs), zonulin, sphingomyelins, acylcarnitines, and trimethylamine N-oxide (TMAO) were found to be increased in hypertensives. Changes in these biomarkers further establish the relation between gut epithelial health and high blood pressure (BP). Participants affected by diseases have an overall lower rate of acquiring new genes, which results in a low richness of genes in them compared with healthy individuals. There is increased expression of the choline utilization (cutC) gene and reduced expression of genes associated with biosynthesis and transport of amino acids in high-BP participants. The unique changes in the composition of the microbiota, functional changes in genes, and metabolome collectively help for a better understanding of the pathogenesis of HTN and also suggest the gut as a promising new therapeutic target for HTN. To establish a further causal relationship between the two, more research is required.
Collapse
Affiliation(s)
- Shaili S Naik
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Surat Municipal Institute of Medical Education and Research (SMIMER) Hospital and Medical College, Surat, IND
| | - Shivana Ramphall
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Research, American University of Antigua, Osbourn, ATG
| | - Swarnima Rijal
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Vishakh Prakash
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Government Medical College Kozhikode, Kozhikode, IND
| | - Heba Ekladios
- Department of Psychiatry, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jiya Mulayamkuzhiyil Saju
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Sree Narayana Institute of Medical Sciences, Ernakulam, IND
- General Surgery, Government Medical College, Thiruvananthapuram, Trivandrum, IND
| | - Naishal Mandal
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Surat Municipal Institute of Medical Education and Research (SMIMER) Hospital and Medical College, Surat, IND
| | - Nang I Kham
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rabia Shahid
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sathish Venugopal
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
6
|
Roslund MI, Parajuli A, Hui N, Puhakka R, Grönroos M, Soininen L, Nurminen N, Oikarinen S, Cinek O, Kramná L, Schroderus AM, Laitinen OH, Kinnunen T, Hyöty H, Sinkkonen A. A Placebo-controlled double-blinded test of the biodiversity hypothesis of immune-mediated diseases: Environmental microbial diversity elicits changes in cytokines and increase in T regulatory cells in young children. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113900. [PMID: 35930838 DOI: 10.1016/j.ecoenv.2022.113900] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND According to the biodiversity hypothesis of immune-mediated diseases, lack of microbiological diversity in the everyday living environment is a core reason for dysregulation of immune tolerance and - eventually - the epidemic of immune-mediated diseases in western urban populations. Despite years of intense research, the hypothesis was never tested in a double-blinded and placebo-controlled intervention trial. OBJECTIVE We aimed to perform the first placebo-controlled double-blinded test that investigates the effect of biodiversity on immune tolerance. METHODS In the intervention group, children aged 3-5 years were exposed to playground sand enriched with microbially diverse soil, or in the placebo group, visually similar, but microbially poor sand colored with peat (13 participants per treatment group). Children played twice a day for 20 min in the sandbox for 14 days. Sand, skin and gut bacterial, and blood samples were taken at baseline and after 14 days. Bacterial changes were followed for 28 days. Sand, skin and gut metagenome was determined by high throughput sequencing of bacterial 16 S rRNA gene. Cytokines were measured from plasma and the frequency of blood regulatory T cells was defined as a percentage of total CD3 +CD4 + T cells. RESULTS Bacterial richness (P < 0.001) and diversity (P < 0.05) were higher in the intervention than placebo sand. Skin bacterial community, including Gammaproteobacteria, shifted only in the intervention treatment to resemble the bacterial community in the enriched sand (P < 0.01). Mean change in plasma interleukin-10 (IL-10) concentration and IL-10 to IL-17A ratio supported immunoregulation in the intervention treatment compared to the placebo treatment (P = 0.02). IL-10 levels (P = 0.001) and IL-10 to IL-17A ratio (P = 0.02) were associated with Gammaproteobacterial community on the skin. The change in Treg frequencies was associated with the relative abundance of skin Thermoactinomycetaceae 1 (P = 0.002) and unclassified Alphaproteobacteria (P < 0.001). After 28 days, skin bacterial community still differed in the intervention treatment compared to baseline (P < 0.02). CONCLUSIONS This is the first double-blinded placebo-controlled study to show that daily exposure to microbial biodiversity is associated with immune modulation in humans. The findings support the biodiversity hypothesis of immune-mediated diseases. We conclude that environmental microbiota may contribute to child health, and that adding microbiological diversity to everyday living environment may support immunoregulation.
Collapse
Affiliation(s)
- Marja I Roslund
- Natural Resources Institute Finland (Luke), Helsinki and Turku, Finland; Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Anirudra Parajuli
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Nan Hui
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Mira Grönroos
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Laura Soininen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Noora Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Ondřej Cinek
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University, V Úvalu 84, Praha 5, 150 06 Prague, Czech Republic
| | - Lenka Kramná
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University, V Úvalu 84, Praha 5, 150 06 Prague, Czech Republic
| | - Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Olli H Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Eastern Finland Laboratory Centre (ISLAB), Kuopio, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland; Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Aki Sinkkonen
- Natural Resources Institute Finland (Luke), Helsinki and Turku, Finland.
| |
Collapse
|
7
|
Sahoyama Y, Hamazato F, Shiozawa M, Nakagawa T, Suda W, Ogata Y, Hachiya T, Kawakami E, Hattori M. Multiple nutritional and gut microbial factors associated with allergic rhinitis: the Hitachi Health Study. Sci Rep 2022; 12:3359. [PMID: 35233003 PMCID: PMC8888718 DOI: 10.1038/s41598-022-07398-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/15/2022] [Indexed: 11/28/2022] Open
Abstract
Several studies suggest the involvement of dietary habits and gut microbiome in allergic diseases. However, little is known about the nutritional and gut microbial factors associated with the risk of allergic rhinitis (AR). We recruited 186 participants with symptoms of AR and 106 control subjects without symptoms of AR at the Hitachi Health Care Center, Japan. The habitual consumption of 42 selected nutrients were examined using the brief-type self-administered diet history questionnaire. Faecal samples were collected and subjected to amplicon sequencing of the 16S ribosomal RNA gene hypervariable regions. Association analysis revealed that four nutrients (retinol, vitamin A, cryptoxanthin, and copper) were negatively associated with AR. Among 40 genera examined, relative abundance of Prevotella and Escherichia were associated with AR. Furthermore, significant statistical interactions were observed between retinol and Prevotella. The age- and sex-adjusted odds of AR were 25-fold lower in subjects with high retinol intake and high Prevotella abundance compared to subjects with low retinol intake and low Prevotella abundance. Our data provide insights into complex interplay between dietary nutrients, gut microbiome, and the development of AR.
Collapse
Affiliation(s)
- Yukari Sahoyama
- Technology Innovation Div., Hitachi High-Tech Corporation, Business Tower, Toranomon Hills, 1-17-1 Minato-ku, Toranomon, Tokyo, 105-6409, Japan.
| | - Fumiaki Hamazato
- Technology Innovation Div., Hitachi High-Tech Corporation, Business Tower, Toranomon Hills, 1-17-1 Minato-ku, Toranomon, Tokyo, 105-6409, Japan
| | - Manabu Shiozawa
- Technology Innovation Div., Hitachi High-Tech Corporation, Business Tower, Toranomon Hills, 1-17-1 Minato-ku, Toranomon, Tokyo, 105-6409, Japan
| | - Tohru Nakagawa
- Hitachi Health Care Center, Hitachi Ltd., Ibaraki, Japan
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yusuke Ogata
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Eiryo Kawakami
- Medical Sciences Innovation Hub Program, RIKEN, Yokohama, Japan.,Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masahira Hattori
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
8
|
Yang Y, Jiang X, Cai X, Zhang L, Li W, Che L, Fang Z, Feng B, Lin Y, Xu S, Li J, Zhao X, Wu D, Zhuo Y. Deprivation of Dietary Fiber Enhances Susceptibility of Piglets to Lung Immune Stress. Front Nutr 2022; 9:827509. [PMID: 35223957 PMCID: PMC8867169 DOI: 10.3389/fnut.2022.827509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Growing evidence suggests that dietary fiber enhances short-chain fatty acid (SCFA) producing gut microbes, improving lung immunity against invading pathogens via the gut–lung axis. This study investigated the effects of dietary fiber on lung immune stress after challenge with complete Freund's adjuvant (CFA) containing killed Mycobacterium tuberculosis. Thirty-six healthy hybrid Duroc, Landrace, and Yorkshire male piglets (9.7 ± 1.07 kg, 35 ± 3 days) were randomly fed a low fiber (LF) diet formulated with semipurified corn starch, soy protein concentrate, and fishmeal or a high fiber (HF) diet composed of 1,000 g LF diet plus 20 g inulin, and 100 g cellulose. Piglets were housed individually in the metabolism cages with eighteen replicates per group, with one pig per cage. All the piglets received similar levels of digestible energy and lysine and had similar weight gain. After dietary treatment for 28 days, nine piglets per group were intravenously administered CFA (0.4 mg/kg) or an equivalent amount of sterile saline in a 2 × 2 factorial arrangement. In piglets fed the LF diet, CFA caused lung damage and elevated serum C-reactive protein and relative mRNA expression of genes related to lung inflammation (NLRP3, Casp1, ASC, IL1β, IL18, Bax). Compared with the LF diet, the HF diet increased bacterial diversity and Deferribacteres (p = 0.01) in the phylum level and unidentified_Ruminococcaceae (p = 0.03) and Catenisphaera (p < 0.01) in the genus level. The HF diet improved increased short-chain fatty acids in feces, blood, cecal, and colonic digesta; reduced lung damage; and promoted lung recovery. Overall, dietary fiber deprivation enhanced the risk of piglets to lung immune stress, demonstrating the importance of dietary fiber in gut–lung health.
Collapse
Affiliation(s)
- Yi Yang
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Jiang
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xuelin Cai
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Lijia Zhang
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Wentao Li
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Lianqiang Che
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhengfeng Fang
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bin Feng
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yan Lin
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shengyu Xu
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jian Li
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xilun Zhao
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - De Wu
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yong Zhuo
- State Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
9
|
McDonald CR, Weckman AM, Wright JK, Conroy AL, Kain KC. Developmental origins of disease highlight the immediate need for expanded access to comprehensive prenatal care. Front Public Health 2022; 10:1021901. [PMID: 36504964 PMCID: PMC9730730 DOI: 10.3389/fpubh.2022.1021901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
The prenatal environment plays a critical role in shaping fetal development and ultimately the long-term health of the child. Here, we present data linking prenatal health, via maternal nutrition, comorbidities in pregnancy (e.g., diabetes, hypertension), and infectious and inflammatory exposures, to lifelong health through the developmental origins of disease framework. It is well-established that poor maternal health puts a child at risk for adverse outcomes in the first 1,000 days of life, yet the full health impact of the in utero environment is not confined to this narrow window. The developmental origins of disease framework identifies cognitive, neuropsychiatric, metabolic and cardiovascular disorders, and chronic diseases in childhood and adulthood that have their genesis in prenatal life. This perspective highlights the enormous public health implications for millions of pregnancies where maternal care, and therefore maternal health and fetal health, is lacking. Despite near universal agreement that access to antenatal care is a priority to protect the health of women and children in the first 1,000 days of life, insufficient progress has been achieved. Instead, in some regions there has been a political shift toward deprioritizing maternal health, which will further negatively impact the health and safety of pregnant people and their children across the lifespan. In this article we argue that the lifelong health impact attributed to the perinatal environment justifies policies aimed at improving access to comprehensive antenatal care globally.
Collapse
Affiliation(s)
- Chloe R McDonald
- Sandra A. Rotman (SAR) Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Andrea M Weckman
- Sandra A. Rotman (SAR) Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Julie K Wright
- Sandra A. Rotman (SAR) Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada.,Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrea L Conroy
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Kevin C Kain
- Sandra A. Rotman (SAR) Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada.,Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.,Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Aganetti MA, Cruz CS, Galvão I, Engels DF, Ricci MF, Vieira AT. The Gut Microbiota and Immunopathophysiology. COMPREHENSIVE PHARMACOLOGY 2022:492-514. [DOI: 10.1016/b978-0-12-820472-6.00128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
11
|
Interplay between Candida albicans and Lactic Acid Bacteria in the Gastrointestinal Tract: Impact on Colonization Resistance, Microbial Carriage, Opportunistic Infection, and Host Immunity. Clin Microbiol Rev 2021; 34:e0032320. [PMID: 34259567 PMCID: PMC8404691 DOI: 10.1128/cmr.00323-20] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Emerging studies have highlighted the disproportionate role of Candida albicans in influencing both early community assembly of the bacterial microbiome and dysbiosis during allergic diseases and intestinal inflammation. Nonpathogenic colonization of the human gastrointestinal (GI) tract by C. albicans is common, and the role of this single fungal species in modulating bacterial community reassembly after broad-spectrum antibiotics can be readily recapitulated in mouse studies. One of the most notable features of C. albicans-associated dysbiotic states is a marked change in the levels of lactic acid bacteria (LAB). C. albicans and LAB share metabolic niches throughout the GI tract, and in vitro studies have identified various interactions between these microbes. The two predominant LAB affected are Lactobacillus species and Enterococcus species. Lactobacilli can antagonize enterococci and C. albicans, while Enterococcus faecalis and C. albicans have been reported to exhibit a mutualistic relationship. E. faecalis and C. albicans are also causative agents of a variety of life-threatening infections, are frequently isolated together from mixed-species infections, and share certain similarities in clinical presentation-most notably their emergence as opportunistic pathogens following disruption of the microbiota. In this review, we discuss and model the mechanisms used by Lactobacillus species, E. faecalis, and C. albicans to modulate each other's growth and virulence in the GI tract. With multidrug-resistant E. faecalis and C. albicans strains becoming increasingly common in hospital settings, examining the interplay between these three microbes may provide novel insights for enhancing the efficacy of existing antimicrobial therapies.
Collapse
|
12
|
Vithoulkas G. An integrated perspective on transmutation of acute inflammation into chronic and the role of the microbiome. J Med Life 2021; 14:740-747. [PMID: 35126742 PMCID: PMC8811668 DOI: 10.25122/jml-2021-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
The Continuum theory and the Levels of Health theory were separately proposed to explain the myriad responses to treatment and understand the process of health and disease in an individual. In light of accumulating evidence on the intricate relationship between the human immune system and microbiome, an attempt is made in this article to connect these two theories to explain the transmutation of the efficiently responding immune system (through the acute inflammatory response and high fever) to one involved in a low-grade chronic inflammatory process (resulting in chronic disease). There is already enough evidence to demonstrate the role of the microbiome in all chronic inflammatory diseases. In this article, we discuss the mechanism by which subjecting a healthy person to continuous drug treatment for acute inflammatory conditions (at a certain time) leads to transmutation to chronic disease. Although this hypothesis requires further experimental evidence, it calls for a reconsideration of the manner in which we treat acute infectious diseases in the population.
Collapse
Affiliation(s)
- George Vithoulkas
- University of the Aegean, Syros, Greece
- Postgraduate Doctors’ Training Institute, Health Care Ministry of the Chuvash Republic, Cheboksary, Russian Federation
| |
Collapse
|
13
|
Gomez MV, Dutta M, Suvorov A, Shi X, Gu H, Mani S, Yue Cui J. Early Life Exposure to Environmental Contaminants (BDE-47, TBBPA, and BPS) Produced Persistent Alterations in Fecal Microbiome in Adult Male Mice. Toxicol Sci 2021; 179:14-30. [PMID: 33078840 DOI: 10.1093/toxsci/kfaa161] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The gut microbiome is a pivotal player in toxicological responses. We investigated the effects of maternal exposure to 3 human health-relevant toxicants (BDE-47, tetrabromobisphenol [TBBPA], and bisphenol S [BPS]) on the composition and metabolite levels (bile acids [BAs] and short-chain fatty acids [SCFAs]) of the gut microbiome in adult pups. CD-1 mouse dams were orally exposed to vehicle (corn oil, 10 ml/kg), BDE-47 (0.2 mg/kg), TBBPA (0.2 mg/kg), or BPS (0.2 mg/kg) once daily from gestational day 8 to the end of lactation (postnatal day 21). 16S rRNA sequencing and targeted metabolomics were performed in feces of 20-week-old adult male pups (n = 14 - 23/group). Host gene expression and BA levels were quantified in liver. BPS had the most prominent effect on the beta-diversity of the fecal microbiome compared with TBPPA and BDE-47 (QIIME). Seventy-three taxa were persistently altered by at least 1 chemical, and 12 taxa were commonly regulated by all chemicals (most of which were from the Clostridia class and were decreased). The most distinct microbial biomarkers were S24-7 for BDE-47, Rikenellaceae for TBBPA, and Lactobacillus for BPS (LefSe). The community-wide contributions to the shift in microbial pathways were predicted using FishTaco. Consistent with FishTaco predictions, BDE-47 persistently increased fecal and hepatic BAs within the 12α hydroxylation pathway, corresponding to an up-regulation with the hepatic BA-synthetic enzyme Cyp7a1. Fecal BAs were also persistently up-regulated by TBBPA and BPS (liquid chromatography-mass spectrometry). TBBPA increased propionic acid and succinate, whereas BPS decreased acetic acid (gas chromatography-mass spectrometry). There was a general trend in the hepatic down-regulation of proinflammatory cytokines and the oxidative stress sensor target gene (Nqo1), and a decrease in G6Pdx (the deficiency of which leads to dyslipidemia). In conclusion, maternal exposure to these toxicants persistently modified the gut-liver axis, which may produce an immune-suppressive and dyslipidemia-prone signature later in life.
Collapse
Affiliation(s)
- Matthew V Gomez
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | - Alexander Suvorov
- Environmental Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Xiaojian Shi
- College of Health Solutions, Arizona State University, Tempe, Arizona, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Tempe, Arizona, USA
| | - Sridhar Mani
- Albert Einstein College of Medicine, Bronx, New York, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
Sharma SR, Karim S. Tick Saliva and the Alpha-Gal Syndrome: Finding a Needle in a Haystack. Front Cell Infect Microbiol 2021; 11:680264. [PMID: 34354960 PMCID: PMC8331069 DOI: 10.3389/fcimb.2021.680264] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023] Open
Abstract
Ticks and tick-borne diseases are significant public health concerns. Bioactive molecules in tick saliva facilitate prolonged blood-feeding and transmission of tick-borne pathogens to the vertebrate host. Alpha-gal syndrome (AGS), a newly reported food allergy, is believed to be induced by saliva proteins decorated with a sugar molecule, the oligosaccharide galactose-⍺-1,3-galactose (α-gal). This syndrome is characterized by an IgE antibody-directed hypersensitivity against α-gal. The α-gal antigen was discovered in the salivary glands and saliva of various tick species including, the Lone Star tick (Amblyomma americanum). The underlying immune mechanisms linking tick bites with α-gal-specific IgE production are poorly understood and are crucial to identify and establish novel treatments for this disease. This article reviews the current understanding of AGS and its involvement with tick species.
Collapse
Affiliation(s)
- Surendra Raj Sharma
- Center for Molecular and Cellular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Shahid Karim
- Center for Molecular and Cellular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
15
|
Hemida MBM, Salin S, Vuori KA, Moore R, Anturaniemi J, Rosendahl S, Barrouin-Melo SM, Hielm-Björkman A. Puppyhood diet as a factor in the development of owner-reported allergy/atopy skin signs in adult dogs in Finland. J Vet Intern Med 2021; 35:2374-2383. [PMID: 34258795 PMCID: PMC8478020 DOI: 10.1111/jvim.16211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The increased prevalence of atopic dermatitis (AD) in dogs necessitates research in its disease etiology. OBJECTIVES To explore the association between puppyhood dietary exposures and prevalence of owner-reported allergy/atopy skin signs (AASS) after the age of 1 year. ANIMALS Four thousand and twenty-two dogs were eligible, 1158 cases, and 2864 controls. METHODS This cross-sectional hypothesis-driven observational study was extracted from the DogRisk food frequency questionnaire. Forty-six food items and the ratio of 4 major diet types were tested for their association with AASS incidence later in life. Potential puppyhood dietary risk factors for AASS incidence were specified using binary multivariable logistic regression. The model was adjusted for age and sex. RESULTS Eating raw tripe (odds ratio, 95% confidence intervals OR, 95% CI = 0.36, 0.16-0.79; P = .01), raw organ meats (OR, 95% CI = 0.23, 0.08-0.67; P = .007), human meal leftovers, and fish oil supplements as well as eating more that 20% of the diet as raw and/or <80% of the diet as dry, in general, were associated with significantly lower AASS incidence in adulthood. In contrast, dogs fed fruits (OR, 95% CI = 2.01, 1.31-3.07; P = .001), mixed-oil supplements, dried animal parts, and dogs that drank from puddles showed significantly higher AASS incidence in adulthood. CONCLUSIONS AND CLINICAL IMPORTANCE Puppyhood exposure to raw animal-based foods might have a protective influence on AASS incidence in adulthood, while puppyhood exposure to mixed oils, heat processed foods and sugary fruits might be a potential risk factor of AASS incidence later. The study suggests a causal relationship but does not prove it.
Collapse
Affiliation(s)
- Manal B M Hemida
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Siru Salin
- Department of Agricultural Sciences, Faculty of Agriculture Forestry, University of Helsinki, Helsinki, Finland
| | - Kristiina A Vuori
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Robin Moore
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Anturaniemi
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sarah Rosendahl
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Stella Maria Barrouin-Melo
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Department of Veterinary Anatomy, Pathology and Clinics, School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Anna Hielm-Björkman
- Department of Equine and Small Animal Medicine, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
The role of the microbiome in gastrointestinal inflammation. Biosci Rep 2021; 41:228872. [PMID: 34076695 PMCID: PMC8201460 DOI: 10.1042/bsr20203850] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
The microbiome plays an important role in maintaining human health. Despite multiple factors being attributed to the shaping of the human microbiome, extrinsic factors such diet and use of medications including antibiotics appear to dominate. Mucosal surfaces, particularly in the gut, are highly adapted to be able to tolerate a large population of microorganisms whilst still being able to produce a rapid and effective immune response against infection. The intestinal microbiome is not functionally independent from the host mucosa and can, through presentation of microbe-associated molecular patterns (MAMPs) and generation of microbe-derived metabolites, fundamentally influence mucosal barrier integrity and modulate host immunity. In a healthy gut there is an abundance of beneficial bacteria that help to preserve intestinal homoeostasis, promote protective immune responses, and limit excessive inflammation. The importance of the microbiome is further highlighted during dysbiosis where a loss of this finely balanced microbial population can lead to mucosal barrier dysfunction, aberrant immune responses, and chronic inflammation that increases the risk of disease development. Improvements in our understanding of the microbiome are providing opportunities to harness members of a healthy microbiota to help reverse dysbiosis, reduce inflammation, and ultimately prevent disease progression.
Collapse
|
17
|
Sorensen K, Cawood AL, Gibson GR, Cooke LH, Stratton RJ. Amino Acid Formula Containing Synbiotics in Infants with Cow's Milk Protein Allergy: A Systematic Review and Meta-Analysis. Nutrients 2021; 13:935. [PMID: 33799379 PMCID: PMC7998621 DOI: 10.3390/nu13030935] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cow's milk protein allergy (CMPA) is associated with dysbiosis of the infant gut microbiome, with allergic and immune development implications. Studies show benefits of combining synbiotics with hypoallergenic formulae, although evidence has never been systematically examined. This review identified seven publications of four randomised controlled trials comparing an amino acid formula (AAF) with an AAF containing synbiotics (AAF-Syn) in infants with CMPA (mean age 8.6 months; 68% male, mean intervention 27.3 weeks, n = 410). AAF and AAF-Syn were equally effective in managing allergic symptoms and promoting normal growth. Compared to AAF, significantly fewer infants fed AAF-Syn had infections (OR 0.35 (95% CI 0.19-0.67), p = 0.001). Overall medication use, including antibacterials and antifectives, was lower among infants fed AAF-Syn. Significantly fewer infants had hospital admissions with AAF-Syn compared to AAF (8.8% vs. 20.2%, p = 0.036; 56% reduction), leading to potential cost savings per infant of £164.05-£338.77. AAF-Syn was associated with increased bifidobacteria (difference in means 31.75, 95% CI 26.04-37.45, p < 0.0001); reduced Eubacterium rectale and Clostridium coccoides (difference in means -19.06, 95% CI -23.15 to -14.97, p < 0.0001); and reduced microbial diversity (p < 0.05), similar to that described in healthy breastfed infants, and may be associated with the improved clinical outcomes described. This review provides evidence that suggests combining synbiotics with AAF produces clinical benefits with potential economic implications.
Collapse
Affiliation(s)
- Katy Sorensen
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
| | - Abbie L. Cawood
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
- Institute of Human Nutrition, Faculty of Medicine, Mailpoint 113, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK; (A.L.C.); (R.J.S.)
| | - Glenn R. Gibson
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AP, UK;
| | - Lisa H. Cooke
- Department of Nutrition and Dietetics, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol BS2 8BJ, UK;
| | - Rebecca J. Stratton
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
- Institute of Human Nutrition, Faculty of Medicine, Mailpoint 113, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK; (A.L.C.); (R.J.S.)
| |
Collapse
|
18
|
Roslund MI, Puhakka R, Grönroos M, Nurminen N, Oikarinen S, Gazali AM, Cinek O, Kramná L, Siter N, Vari HK, Soininen L, Parajuli A, Rajaniemi J, Kinnunen T, Laitinen OH, Hyöty H, Sinkkonen A. Biodiversity intervention enhances immune regulation and health-associated commensal microbiota among daycare children. SCIENCE ADVANCES 2020; 6:eaba2578. [PMID: 33055153 PMCID: PMC7556828 DOI: 10.1126/sciadv.aba2578] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 08/14/2020] [Indexed: 05/02/2023]
Abstract
As the incidence of immune-mediated diseases has increased rapidly in developed societies, there is an unmet need for novel prophylactic practices to fight against these maladies. This study is the first human intervention trial in which urban environmental biodiversity was manipulated to examine its effects on the commensal microbiome and immunoregulation in children. We analyzed changes in the skin and gut microbiota and blood immune markers of children during a 28-day biodiversity intervention. Children in standard urban and nature-oriented daycare centers were analyzed for comparison. The intervention diversified both the environmental and skin Gammaproteobacterial communities, which, in turn, were associated with increases in plasma TGF-β1 levels and the proportion of regulatory T cells. The plasma IL-10:IL-17A ratio increased among intervention children during the trial. Our findings suggest that biodiversity intervention enhances immunoregulatory pathways and provide an incentive for future prophylactic approaches to reduce the risk of immune-mediated diseases in urban societies.
Collapse
Affiliation(s)
- Marja I Roslund
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Mira Grönroos
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Noora Nurminen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Ahmad M Gazali
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ondřej Cinek
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, V Úvalu 84, Praha 5, 150 06 Prague, Czech Republic
| | - Lenka Kramná
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and University Hospital Motol, V Úvalu 84, Praha 5, 150 06 Prague, Czech Republic
| | - Nathan Siter
- Faculty of Built Environment, Tampere University, Korkeakoulunkatu 5, FI-33720 Tampere, Finland
| | - Heli K Vari
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Laura Soininen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Anirudra Parajuli
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland
| | - Juho Rajaniemi
- Faculty of Built Environment, Tampere University, Korkeakoulunkatu 5, FI-33720 Tampere, Finland
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Eastern Finland Laboratory Centre (ISLAB), Kuopio, Finland
| | - Olli H Laitinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Aki Sinkkonen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Niemenkatu 73, FI-15140 Lahti, Finland.
- Natural Resources Institute Finland Luke, Itäinen Pitkäkatu 4A, 20520 Turku, Finland
| |
Collapse
|
19
|
Developmental Origins of Health and Disease: Impact of environmental dust exposure in modulating microbiome and its association with non-communicable diseases. J Dev Orig Health Dis 2020; 11:545-556. [PMID: 32536356 DOI: 10.1017/s2040174420000549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Non-communicable diseases (NCDs) including obesity, diabetes, and allergy are chronic, multi-factorial conditions that are affected by both genetic and environmental factors. Over the last decade, the microbiome has emerged as a possible contributor to the pathogenesis of NCDs. Microbiome profiles were altered in patients with NCDs, and shift in microbial communities was associated with improvement in these health conditions. Since the genetic component of these diseases cannot be altered, the ability to manipulate the microbiome holds great promise for design of novel therapies in the prevention and treatment of NCDs. Together, the Developmental Origins of Health and Disease concept and the microbial hypothesis propose that early life exposure to environmental stimuli will alter the development and composition of the human microbiome, resulting in health consequences. Recent studies indicated that the environment we are exposed to in early life is instrumental in shaping robust immune development, possibly through modulation of the human microbiome (skin, airway, and gut). Despite much research into human microbiome, the origin of their constituent microbiota remains unclear. Dust (also known as particulate matter) is a key determinant of poor air quality in the modern urban environment. It is ubiquitous and serves as a major source and reservoir of microbial communities that modulates the human microbiome, contributing to health and disease. There are evidence that reported significant associations between environmental dust and NCDs. In this review, we will focus on the impact of dust exposure in shaping the human microbiome and its possible contribution to the development of NCDs.
Collapse
|
20
|
Sbihi H, Boutin RCT, Cutler C, Suen M, Finlay BB, Turvey SE. Thinking bigger: How early-life environmental exposures shape the gut microbiome and influence the development of asthma and allergic disease. Allergy 2019; 74:2103-2115. [PMID: 30964945 DOI: 10.1111/all.13812] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/27/2019] [Accepted: 03/22/2019] [Indexed: 02/06/2023]
Abstract
Imbalance, or dysbiosis, of the gut microbiome of infants has been linked to an increased risk of asthma and allergic diseases. Most studies to date have provided a wealth of data showing correlations between early-life risk factors for disease and changes in the structure of the gut microbiome that disrupt normal immunoregulation. These studies have typically focused on one specific risk factor, such as mode of delivery or early-life antibiotic use. Such "micro-level" exposures have a considerable impact on affected individuals but not necessarily the whole population. In this review, we place these mechanisms under a larger lens that takes into account the influence of upstream "macro-level" environmental factors such as air pollution and the built environment. While these exposures likely have a smaller impact on the microbiome at an individual level, their ubiquitous nature confers them with a large influence at the population level. We focus on features of the indoor and outdoor human-made environment, their microbiomes and the research challenges inherent in integrating the built environment microbiomes with the early-life gut microbiome. We argue that an exposome perspective integrating internal and external microbiomes with macro-level environmental factors can provide a more comprehensive framework to define how environmental exposures can shape the gut microbiome and influence the development of allergic disease.
Collapse
Affiliation(s)
- Hind Sbihi
- Department of Pediatrics, British Columbia Children’s Hospital The University of British Columbia Vancouver British Columbia Canada
| | - Rozlyn CT. Boutin
- Department of Microbiology and Immunology, Michael Smith Laboratories The University of British Columbia Vancouver British Columbia Canada
| | - Chelsea Cutler
- Department of Pediatrics, British Columbia Children’s Hospital The University of British Columbia Vancouver British Columbia Canada
| | - Mandy Suen
- Department of Pediatrics, British Columbia Children’s Hospital The University of British Columbia Vancouver British Columbia Canada
| | - B. Brett Finlay
- Department of Microbiology and Immunology, Michael Smith Laboratories The University of British Columbia Vancouver British Columbia Canada
| | - Stuart E. Turvey
- Department of Pediatrics, British Columbia Children’s Hospital The University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
21
|
García-Rivero JL. The Microbiome and Asthma. Arch Bronconeumol 2019; 56:1-2. [PMID: 30803838 DOI: 10.1016/j.arbres.2019.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 11/29/2022]
|
22
|
Bundgaard-Nielsen C, Hagstrøm S, Sørensen S. Interpersonal Variations in Gut Microbiota Profiles Supersedes the Effects of Differing Fecal Storage Conditions. Sci Rep 2018; 8:17367. [PMID: 30478355 PMCID: PMC6255890 DOI: 10.1038/s41598-018-35843-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
Due to ease of acquisition, fecal samples are often used in studies investigating gut microbiota. Improper handling of these samples can lead to bacterial growth and alter bacterial composition. While freezing samples at −80 °C is considered gold standard, this is not suitable for studies utilizing self-sampling by lay participants or field studies. Thus to effectively prevent bacterial growth, techniques that allow efficient fecal storage outside laboratory facilities are needed. Fecal samples were collected from three donors. From each donor feces, 45 samples were collected and stored either freshly frozen at −80 or −20 °C, or in three separate storage buffers at room temperature or 4 °C for 24 or 72 hours. Bacterial composition was analyzed using Illumina amplicon sequencing of the V4 region of the 16 S rRNA gene. While storage conditions did affect bacterial composition and diversity compared to storage at −80 °C, the variation between donors superseded the variations introduced by storage. Samples stored at −20 °C most closely resembled those stored at −80 °C. When investigating variations in bacterial composition between separate study populations, fecal samples can efficiently be stored in −20 °C freezers or in one of the presented storage buffers, without severe alterations in bacterial composition.
Collapse
Affiliation(s)
- Caspar Bundgaard-Nielsen
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Søren Hagstrøm
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Department of Pediatrics, Aalborg University Hospital, Aalborg, Denmark
| | - Suzette Sørensen
- Centre for Clinical Research, North Denmark Regional Hospital, Hjørring, Denmark. .,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
23
|
Microbial and Nutritional Programming-The Importance of the Microbiome and Early Exposure to Potential Food Allergens in the Development of Allergies. Nutrients 2018; 10:nu10101541. [PMID: 30340391 PMCID: PMC6212882 DOI: 10.3390/nu10101541] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
The “microbiota hypothesis” ties the increase in allergy rates observed in highly developed countries over the last decades to disturbances in the gut microbiota. Gut microbiota formation depends on a number of factors and occurs over approximately 1000 days of life, including the prenatal period. During this period the microbiota helps establish the functional immune phenotype, including immune tolerance. The development of immune tolerance depends also on early exposure to potential food allergens, a process referred to as nutritional programming. This article elaborates on the concepts of microbial and nutritional programming and their role in the primary prevention of allergy.
Collapse
|
24
|
Bao HD, Pang MD, Olaniran A, Zhang XH, Zhang H, Zhou Y, Sun LC, Schmidt S, Wang R. Alterations in the diversity and composition of mice gut microbiota by lytic or temperate gut phage treatment. Appl Microbiol Biotechnol 2018; 102:10219-10230. [PMID: 30302521 DOI: 10.1007/s00253-018-9378-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/29/2018] [Accepted: 09/05/2018] [Indexed: 12/12/2022]
Abstract
Phages, the most abundant species in the mammalian gut, have numerous advantages as biocontrol agent over antibiotics. In this study, mice were orally treated with the lytic gut phage PA13076 (group B), the temperate phage BP96115 (group C), no phage (group A), or streptomycin (group D) over 31 days. At the end of the experiment, fecal microbiota diversity and composition was determined and compared using high-throughput sequencing of the V3-V4 hyper-variable region of the 16S rRNA gene and virus-like particles (VLPs) were quantified in feces. There was high diversity and richness of microbiota in the lytic and temperate gut phage-treated mice, with the lytic gut phage causing an increased alpha diversity based on the Chao1 index (p < 0.01). However, the streptomycin treatment reduced the microbiota diversity and richness (p = 0.0299). Both phage and streptomycin treatments reduced the abundance of Bacteroidetes at the phylum level (p < 0.01) and increased the abundance of the phylum Firmicutes. Interestingly, two beneficial genera, Lactobacillus and Bifidobacterium, were enhanced by treatment with the lytic and temperate gut phage. The abundance of the genus Escherichia/Shigella was higher in mice after temperate phage administration than in the control group (p < 0.01), but lower than in the streptomycin group. Moreover, streptomycin treatment increased the abundance of the genera Klebsiella and Escherichia/Shigella (p < 0.01). In terms of the gut virome, fecal VLPs did not change significantly after phage treatment. This study showed that lytic and temperate gut phage treatment modulated the composition and diversity of gut microbiota and the lytic gut phage promoted a beneficial gut ecosystem, while the temperate phage may promote conditions enabling diseases to occur.
Collapse
Affiliation(s)
- Hong-Duo Bao
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, China
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg Campus, Private Bag X01, Pietermaritzburg, 3201, South Africa
| | - Mao-da Pang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Ademola Olaniran
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - Xu-Hui Zhang
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, China
| | - Hui Zhang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Yan Zhou
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Li-Chang Sun
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Stefan Schmidt
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg Campus, Private Bag X01, Pietermaritzburg, 3201, South Africa.
| | - Ran Wang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China.
| |
Collapse
|
25
|
Microbiota Composition in Upper Respiratory Tracts of Healthy Children in Shenzhen, China, Differed with Respiratory Sites and Ages. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6515670. [PMID: 30013985 PMCID: PMC6022278 DOI: 10.1155/2018/6515670] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
The upper respiratory tract (URT) is home to various microbial commensals, which function as competitors to pathogens and help train the immune system. However, few studies have reported the normal microbiota carriage in the URT of healthy Chinese children. In this study, we performed a 16S rDNA gene sequencing analysis of 83 anterior nares (ANs), 60 nasopharynx (NP), and 97 oropharynx (OP) samples from 98 healthy children in Shenzhen, China (≤12 years of age). The microbiota in ANs and NP is the same at different ages and typical species in these sites include Moraxella, Staphylococcus, Corynebacterium, Streptococcus, and Dolosigranulum. By contrast, the OP is primarily colonized by Streptococcus, Prevotella, Neisseria, Veillonella, Rothia, Leptotrichia, and Haemophilus. Streptococcus and Rothia keep low abundance in OP microbiota of children ≤1 year old, whereas Prevotella, Neisseria, Haemophilus, and Leptotrichia amass significantly in individuals >1 year old. This work furnishes an important reference for understanding microbial dysbiosis in the URT of Chinese paediatric patients.
Collapse
|
26
|
Stiemsma LT, Michels KB. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics 2018; 141:e20172437. [PMID: 29519955 PMCID: PMC5869344 DOI: 10.1542/peds.2017-2437] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Abstract
Although the prominent role of the microbiome in human health has been established, the early-life microbiome is now being recognized as a major influence on long-term human health and development. Variations in the composition and functional potential of the early-life microbiome are the result of lifestyle factors, such as mode of birth, breastfeeding, diet, and antibiotic usage. In addition, variations in the composition of the early-life microbiome have been associated with specific disease outcomes, such as asthma, obesity, and neurodevelopmental disorders. This points toward this bacterial consortium as a mediator between early lifestyle factors and health and disease. In addition, variations in the microbial intrauterine environment may predispose neonates to specific health outcomes later in life. A role of the microbiome in the Developmental Origins of Health and Disease is supported in this collective research. Highlighting the early-life critical window of susceptibility associated with microbiome development, we discuss infant microbial colonization, beginning with the maternal-to-fetal exchange of microbes in utero and up through the influence of breastfeeding in the first year of life. In addition, we review the available disease-specific evidence pointing toward the microbiome as a mechanistic mediator in the Developmental Origins of Health and Disease.
Collapse
Affiliation(s)
- Leah T Stiemsma
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Karin B Michels
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
27
|
Abstract
Gut dysbiosis is associated with many non-communicable human diseases, but the mechanisms maintaining homeostasis remain incompletely understood. Recent insights suggest that during homeostasis, epithelial hypoxia limits oxygen availability in the colon, thereby maintaining a balanced microbiota that functions as a microbial organ, producing metabolites contributing to host nutrition, immune education and niche protection. Dysbiosis is characterized by a shift in the microbial community structure from obligate to facultative anaerobes, suggesting oxygen as an important ecological driver of microbial organ dysfunction. The ensuing disruption of gut homeostasis can lead to non- communicable disease because microbiota-derived metabolites are either depleted or generated at harmful concentrations. This Opinion article describes the concept that host control over the microbial ecosystem in the colon is critical for the composition and function of our microbial organ, which provides a theoretical framework for linking microorganisms to non-communicable diseases.
Collapse
Affiliation(s)
- Mariana X Byndloss
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616, USA
| |
Collapse
|
28
|
Di Lorenzo G, Di Bona D, Belluzzo F, Macchia L. Immunological and non-immunological mechanisms of allergic diseases in the elderly: biological and clinical characteristics. IMMUNITY & AGEING 2017; 14:23. [PMID: 29296117 PMCID: PMC5738884 DOI: 10.1186/s12979-017-0105-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/07/2017] [Indexed: 02/08/2023]
Abstract
A better hygiene, a Westernized diet, air pollution, climate changes, and other factors that influence host microbiota, a key player in the induction and maintenance of immunoregulatory circuits and tolerance, are thought to be responsible for the increase of allergic diseases observed in the last years. The increase of allergic diseases in elderly is related to the presence of other factors as several comorbidities that should interfere with the development and the type of allergic reactions. A central role is played by immunosenescence responsible for modifying response to microbiota and triggering inflamm-ageing. In addition, in elderly there is a shift from Th1 responses vs. Th2, hence favouring allergic responses. Better understanding of the mechanisms of immunosenescence and its effects on allergic inflammation will most certainly lead to improved therapy.
Collapse
Affiliation(s)
- Gabriele Di Lorenzo
- Dipartimento BioMedico di Medicina Interna e Specialistica (Di.Bi.M.I.S.), Università di Palermo, Palermo, Italy.,Dipartimento BioMedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), Via del Vespro, 141, 90127 Palermo, Italy
| | - Danilo Di Bona
- Department of Allergy, Clinical Immunology, Emergency Medicine, and Transplants, University of Bari, Bari, Italy
| | - Federica Belluzzo
- Dipartimento BioMedico di Medicina Interna e Specialistica (Di.Bi.M.I.S.), Università di Palermo, Palermo, Italy
| | - Luigi Macchia
- Department of Allergy, Clinical Immunology, Emergency Medicine, and Transplants, University of Bari, Bari, Italy
| |
Collapse
|
29
|
Abstract
This review summarizes the current state of play with regard to food allergy prevention. Food allergy prevention strategies focused on promoting timely introduction of allergenic foods (predominantly peanut) into the infant diet have recently been introduced in several countries. Additional prevention strategies currently under investigation include optimizing infant vitamin D levels, modulating the gut microbiota through use of probiotics, and preventing eczema to reduce the risk of food sensitization through a damaged skin barrier.
Collapse
Affiliation(s)
- Jennifer J Koplin
- Centre of Food and Allergy Research, Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Victoria 3052, Australia
| | - Rachel L Peters
- Centre of Food and Allergy Research, Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Victoria 3052, Australia
| | - Katrina J Allen
- Centre of Food and Allergy Research, Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Allergy and Clinical Immunology, Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Gastroenterology and Clinical Nutrition, Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Institute of Inflammation and Repair, University of Manchester, Oxford Road, Manchester M13 9PL, UK.
| |
Collapse
|
30
|
Presti RM, Flores SC, Palmer BE, Atkinson JJ, Lesko CR, Lau B, Fontenot AP, Roman J, McDyer JF, Twigg HL. Mechanisms Underlying HIV-Associated Noninfectious Lung Disease. Chest 2017; 152:1053-1060. [PMID: 28427967 DOI: 10.1016/j.chest.2017.04.154] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/28/2017] [Accepted: 04/05/2017] [Indexed: 01/15/2023] Open
Abstract
Pulmonary disease remains a primary source of morbidity and mortality in persons living with HIV (PLWH), although the advent of potent combination antiretroviral therapy has resulted in a shift from predominantly infectious to noninfectious pulmonary complications. PLWH are at high risk for COPD, pulmonary hypertension, and lung cancer even in the era of combination antiretroviral therapy. The underlying mechanisms of this are incompletely understood, but recent research in both human and animal models suggests that oxidative stress, expression of matrix metalloproteinases, and genetic instability may result in lung damage, which predisposes PLWH to these conditions. Some of the factors that drive these processes include tobacco and other substance use, direct HIV infection and expression of specific HIV proteins, inflammation, and shifts in the microbiome toward pathogenic and opportunistic organisms. Further studies are needed to understand the relative importance of these factors to the development of lung disease in PLWH.
Collapse
Affiliation(s)
- Rachel M Presti
- Department of Medicine, Washington University School of Medicine, St. Louis, MO.
| | - Sonia C Flores
- Department of Medicine, University of Colorado Denver, Aurora, CO
| | - Brent E Palmer
- Department of Medicine, University of Colorado Denver, Aurora, CO
| | - Jeffrey J Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Catherine R Lesko
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Bryan Lau
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, School of Medicine, Johns Hopkins University, Baltimore, MD
| | | | - Jesse Roman
- Department of Medicine, University of Louisville, Health Sciences Center and Robley Rex VA Medical Center, Louisville, KY
| | - John F McDyer
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Homer L Twigg
- Department of Medicine, Indiana University, Indianapolis, IN
| |
Collapse
|
31
|
Stiemsma LT, Turvey SE. Asthma and the microbiome: defining the critical window in early life. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2017; 13:3. [PMID: 28077947 PMCID: PMC5217603 DOI: 10.1186/s13223-016-0173-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 12/11/2016] [Indexed: 12/23/2022]
Abstract
Asthma is a chronic inflammatory immune disorder of the airways affecting one in ten children in westernized countries. The geographical disparity combined with a generational rise in prevalence, emphasizes that changing environmental exposures play a significant role in the etiology of this disease. The microflora hypothesis suggests that early life exposures are disrupting the composition of the microbiota and consequently, promoting immune dysregulation in the form of hypersensitivity disorders. Animal model research supports a role of the microbiota in asthma and atopic disease development. Further, these model systems have identified an early life critical window, during which gut microbial dysbiosis is most influential in promoting hypersensitivity disorders. Until recently this critical window had not been characterized in humans, but now studies suggest that the ideal time to use microbes as preventative treatments or diagnostics for asthma in humans is within the first 100 days of life. This review outlines the major mouse-model and human studies leading to characterization of the early life critical window, emphasizing studies analyzing the intestinal and airway microbiotas in asthma and atopic disease. This research has promising future implications regarding childhood immune health, as ultimately it may be possible to therapeutically administer specific microbes in early life to prevent the development of asthma in children.
Collapse
Affiliation(s)
- Leah T. Stiemsma
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC Canada
- BC Children’s Hospital, Vancouver, BC Canada
| | - Stuart E. Turvey
- BC Children’s Hospital, Vancouver, BC Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC Canada
- Department of Pediatrics, BC Children’s Hospital, 950 West 28th Avenue, Vancouver, BC V5Z 4H4 Canada
| |
Collapse
|
32
|
Martin VJ, Shreffler WG, Yuan Q. Presumed Allergic Proctocolitis Resolves with Probiotic Monotherapy: A Report of 4 Cases. AMERICAN JOURNAL OF CASE REPORTS 2016; 17:621-4. [PMID: 27568925 PMCID: PMC5004981 DOI: 10.12659/ajcr.898490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Case series Patients: — Final Diagnosis: Allergic proctocolitis Symptoms: Hematochezia • fussiness Medication: — Clinical Procedure: — Specialty: Pediatrics and Neonatology
Collapse
Affiliation(s)
- Victoria J Martin
- Department of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Qian Yuan
- Department of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
33
|
di Mauro G, Bernardini R, Barberi S, Capuano A, Correra A, de’ Angelis GL, Iacono ID, de Martino M, Ghiglioni D, Di Mauro D, Giovannini M, Landi M, Marseglia GL, Martelli A, Miniello VL, Peroni D, Sullo LRMG, Terracciano L, Vascone C, Verduci E, Verga MC, Chiappini E. Prevention of food and airway allergy: consensus of the Italian Society of Preventive and Social Paediatrics, the Italian Society of Paediatric Allergy and Immunology, and Italian Society of Pediatrics. World Allergy Organ J 2016; 9:28. [PMID: 27583103 PMCID: PMC4989298 DOI: 10.1186/s40413-016-0111-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 06/07/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Allergic sensitization in children and allergic diseases arising therefrom are increasing for decades. Several interventions, functional foods, pro- and prebiotics, vitamins are proposed for the prevention of allergies and they can't be uncritically adopted. OBJECTIVE This Consensus document was developed by the Italian Society of Preventive and Social Paediatrics and the Italian Society of Paediatric Allergy and Immunology. The aim is to provide updated recommendations regarding allergy prevention in children. METHODS The document has been issued by a multidisciplinary expert panel and it is intended to be mainly directed to primary care paediatricians. It includes 19 questions which have been preliminarily considered relevant by the panel. Relatively to each question, a literature search has been performed, according to the Italian National Guideline Program. Methodology, and a brief summary of the available literature data, has been provided. Many topics have been analyzed including the role of mother's diet restriction, use of breast/formula/hydrolyzed milk; timing of introduction of complementary foods, role (if any) of probiotics, prebiotics, vitamins, exposure to dust mites, animals and to tobacco smoke. RESULTS Some preventive interventions have a strong level of recommendation. (e.g., the dehumidifier to reduce exposure to mite allergens). With regard to other types of intervention, such as the use of partially and extensively hydrolyzed formulas, the document underlines the lack of evidence of effectiveness. No preventive effect of dietary supplementation with polyunsaturated fatty acids, vitamins or minerals has been demonstrated. There is no preventive effect of probiotics on asthma, rhinitis and allergic diseases. It has demonstrated a modest effect, but steady, in the prevention of atopic dermatitis. CONCLUSIONS The recommendations of the Consensus are based on a careful analysis of the evidence available. The lack of evidence of efficacy does not necessarily imply that some interventions may not be effective, but currently they can't be recommended.
Collapse
Affiliation(s)
- Giuseppe di Mauro
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Roberto Bernardini
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Salvatore Barberi
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Annalisa Capuano
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Antonio Correra
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Gian Luigi de’ Angelis
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Iride Dello Iacono
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Maurizio de Martino
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Daniele Ghiglioni
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Dora Di Mauro
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Marcello Giovannini
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Massimo Landi
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Gian Luigi Marseglia
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Alberto Martelli
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Vito Leonardo Miniello
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Diego Peroni
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Lucilla Ricottini Maria Giuseppa Sullo
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Luigi Terracciano
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Cristina Vascone
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Elvira Verduci
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Maria Carmen Verga
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| | - Elena Chiappini
- Department of Sciences for Health Sciences, Anna Meyer Children’s University Hospital, University of Florence, Viale Pieraccini, 24, Florence, 50100 Italy
| |
Collapse
|
34
|
Antibiotic prescription and food allergy in young children. Allergy Asthma Clin Immunol 2016; 12:41. [PMID: 27536320 PMCID: PMC4988015 DOI: 10.1186/s13223-016-0148-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022] Open
Abstract
Background To assess the relationship between any systemic antibiotic prescription within the first year of life and the presence of an ICD-9-CM diagnosis code for food allergy (FA). Methods This was a matched case–control study conducted using South Carolina Medicaid administrative data. FA cases born between 2007 and 2009 were matched to controls without FA on birth month/year, sex, race/ethnicity. Conditional logistic regression was used to model the adjusted odds ratio (aOR) of FA diagnosis. All models were adjusted for presence of asthma, wheeze, or atopic dermatitis. Results A total of 1504 cases and 5995 controls were identified. Receipt of an antibiotic prescription within the initial 12 months of life was associated with FA diagnosis in unadjusted and adjusted models (aOR 1.21; 95 % CI 1.06–1.39). Compared to children with no antibiotic prescriptions, a linear increase in the aOR was seen with increasing antibiotic prescriptions. Children receiving five or more (aOR 1.64; 95 % CI 1.31–2.05) antibiotic prescriptions were significantly associated with FA diagnosis. The strongest association was noted among recipients of cephalosporin and sulfonamide antibiotics in both unadjusted and adjusted models. Conclusions Receipt of antibiotic prescription in the first year of life is associated with FA diagnosis code in young children after controlling for common covariates. Multiple antibiotic prescriptions are more strongly associated with increases in the odds of FA diagnosis.
Collapse
|
35
|
Prenatal and early-life predictors of atopy and allergic disease in Canadian children: results of the Family Atherosclerosis Monitoring In earLY life (FAMILY) Study. J Dev Orig Health Dis 2016; 7:665-671. [DOI: 10.1017/s2040174416000386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prenatal and early-life environmental exposures play a key role in the development of atopy and allergic disease. The Family Atherosclerosis Monitoring In earLY life Study is a general, population-based Canadian birth cohort that prospectively evaluated prenatal and early-life traits and their association with atopy and/or allergic disease. The study population included 901 babies, 857 mothers and 530 fathers. Prenatal and postnatal risk factors were evaluated through questionnaires collected during the antenatal period and at 1 year. The end points of atopy and allergic diseases in infants were evaluated through questionnaires and skin prick testing. Key outcomes included atopy (24.5%), food allergy (17.5%), cow’s milk allergy (4.8%), wheezing (18.6%) and eczema (16%). The association between infant antibiotic exposure [odds ratio (OR): 2.04, 95% confidence interval (CI): 1.45–2.88] and increased atopy was noted in the multivariate analysis, whereas prenatal maternal exposure to dogs (OR: 0.60, 95% CI: 0.42–0.84) and acetaminophen (OR: 0.68, 95% CI: 0.51–0.92) was associated with decreased atopy. This population-based birth cohort in Canada demonstrated high rates of atopy, food allergy, wheezing and eczema. Several previously reported and some novel prenatal and postnatal exposures were associated with atopy and allergic diseases at 1 year of age.
Collapse
|
36
|
Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-Doutsch S, Kuzeljevic B, Gold MJ, Britton HM, Lefebvre DL, Subbarao P, Mandhane P, Becker A, McNagny KM, Sears MR, Kollmann T, Mohn WW, Turvey SE, Finlay BB. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci Transl Med 2016; 7:307ra152. [PMID: 26424567 DOI: 10.1126/scitranslmed.aab2271] [Citation(s) in RCA: 1163] [Impact Index Per Article: 129.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Asthma is the most prevalent pediatric chronic disease and affects more than 300 million people worldwide. Recent evidence in mice has identified a "critical window" early in life where gut microbial changes (dysbiosis) are most influential in experimental asthma. However, current research has yet to establish whether these changes precede or are involved in human asthma. We compared the gut microbiota of 319 subjects enrolled in the Canadian Healthy Infant Longitudinal Development (CHILD) Study, and show that infants at risk of asthma exhibited transient gut microbial dysbiosis during the first 100 days of life. The relative abundance of the bacterial genera Lachnospira, Veillonella, Faecalibacterium, and Rothia was significantly decreased in children at risk of asthma. This reduction in bacterial taxa was accompanied by reduced levels of fecal acetate and dysregulation of enterohepatic metabolites. Inoculation of germ-free mice with these four bacterial taxa ameliorated airway inflammation in their adult progeny, demonstrating a causal role of these bacterial taxa in averting asthma development. These results enhance the potential for future microbe-based diagnostics and therapies, potentially in the form of probiotics, to prevent the development of asthma and other related allergic diseases in children.
Collapse
Affiliation(s)
- Marie-Claire Arrieta
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada. Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Leah T Stiemsma
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada. Child & Family Research Institute and BC Children's Hospital, Vancouver, British Columbia V4Z 4H4, Canada
| | - Pedro A Dimitriu
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Lisa Thorson
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Shannon Russell
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada. Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Sophie Yurist-Doutsch
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada. Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Boris Kuzeljevic
- Child & Family Research Institute and BC Children's Hospital, Vancouver, British Columbia V4Z 4H4, Canada
| | - Matthew J Gold
- Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Heidi M Britton
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Diana L Lefebvre
- Department of Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, Toronto, Ontario M5S 2J7, Canada. Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R3, Canada. School of Public Health, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Allan Becker
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | - Kelly M McNagny
- Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Malcolm R Sears
- Department of Medicine, McMaster University, Hamilton, Ontario L8S 4L8, Canada
| | - Tobias Kollmann
- Child & Family Research Institute and BC Children's Hospital, Vancouver, British Columbia V4Z 4H4, Canada. Department of Pediatrics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | | | - William W Mohn
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Stuart E Turvey
- Child & Family Research Institute and BC Children's Hospital, Vancouver, British Columbia V4Z 4H4, Canada. Department of Pediatrics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada. Department of Microbiology & Immunology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada. Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| |
Collapse
|
37
|
Grazul H, Kanda LL, Gondek D. Impact of probiotic supplements on microbiome diversity following antibiotic treatment of mice. Gut Microbes 2016; 7:101-14. [PMID: 26963277 PMCID: PMC4856465 DOI: 10.1080/19490976.2016.1138197] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Shifts in microbial populations of the intestinal tract have been associated with a multitude of nutritional, autoimmune, and infectious diseases. The limited diversity following antibiotic treatments creates a window for opportunistic pathogens, diarrhea, and inflammation as the microbiome repopulates. Depending on the antibiotics used, microbial diversity can take weeks to months to recover. To alleviate this loss of diversity in the intestinal microbiota, supplementation with probiotics has become increasingly popular. However, our understanding of the purported health benefits of these probiotic bacteria and their ability to shape the microbiome is significantly lacking. This study examined the impact of probiotics concurrent with antibiotic treatment or during the recovery phase following antibiotic treatment of mice. We found that probiotics did not appear to colonize the intestine themselves or shift the overall diversity of the intestinal microbiota. However, the probiotic supplementation did significantly change the types of bacteria which were present. In particular, during the recovery phase the probiotic caused a suppression of Enterobacteriaceae outgrowth (Shigella and Escherichia) while promoting a blooming of Firmicutes, particularly from the Anaerotruncus genus. These results indicate that probiotics have a significant capacity to remodel the microbiome of an individual recovering from antibiotic therapy.
Collapse
Affiliation(s)
- Hannah Grazul
- Biology Department, Center for Natural Science, Ithaca College, Ithaca NY
| | - L. Leann Kanda
- Biology Department, Center for Natural Science, Ithaca College, Ithaca NY
| | - David Gondek
- Biology Department, Center for Natural Science, Ithaca College, Ithaca NY
| |
Collapse
|
38
|
Cuello-Garcia CA, Fiocchi A, Pawankar R, Yepes-Nuñez JJ, Morgano GP, Zhang Y, Ahn K, Al-Hammadi S, Agarwal A, Gandhi S, Beyer K, Burks W, Canonica GW, Ebisawa M, Kamenwa R, Lee BW, Li H, Prescott S, Riva JJ, Rosenwasser L, Sampson H, Spigler M, Terracciano L, Vereda A, Waserman S, Schünemann HJ, Brożek JL. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Prebiotics. World Allergy Organ J 2016; 9:10. [PMID: 26962387 PMCID: PMC4772464 DOI: 10.1186/s40413-016-0102-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The prevalence of allergic diseases in infants, whose parents and siblings do not have allergy, is approximately 10 % and reaches 20-30 % in those with an allergic first-degree relative. Intestinal microbiota may modulate immunologic and inflammatory systemic responses and, thus, influence development of sensitization and allergy. Prebiotics - non-digestible oligosaccharides that stimulate growth of probiotic bacteria - have been reported to modulate immune responses and their supplementation has been proposed as a preventive intervention. OBJECTIVE The World Allergy Organization (WAO) convened a guideline panel to develop evidence-based recommendations about the use of prebiotics in the prevention of allergy. METHODS The WAO guideline panel identified the most relevant clinical questions about the use of prebiotics for the prevention of allergy. We performed a systematic review of randomized controlled trials of prebiotics, and reviewed the evidence about patient values and preferences, and resource requirements (up to January 2015, with an update on July 29, 2015). We followed the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach to develop recommendations. RESULTS Based on GRADE evidence to decision frameworks, the WAO guideline panel suggests using prebiotic supplementation in not-exclusively breastfed infants and not using prebiotic supplementation in exclusively breastfed infants. Both recommendations are conditional and based on very low certainty of the evidence. We found no experimental or observational study of prebiotic supplementation in pregnant women or in breastfeeding mothers. Thus, the WAO guideline panel chose not to provide a recommendation about prebiotic supplementation in pregnancy or during breastfeeding, at this time. CONCLUSIONS WAO recommendations about prebiotic supplementation for the prevention of allergy are intended to support parents, clinicians and other health care professionals in their decisions whether or not to use prebiotics for the purpose of preventing allergies in healthy, term infants.
Collapse
Affiliation(s)
- Carlos A. Cuello-Garcia
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Tecnologico de Monterrey School of Medicine, Monterrey, Mexico
| | | | - Ruby Pawankar
- />Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Juan José Yepes-Nuñez
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />University of Antioquia, School of Medicine, Medellín, Colombia
| | - Gian Paolo Morgano
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
| | - Yuan Zhang
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
| | - Kangmo Ahn
- />Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Suleiman Al-Hammadi
- />Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Arnav Agarwal
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Shreyas Gandhi
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | | | - Wesley Burks
- />Department of Pediatrics, University of North Carolina, Chapel Hill, NC USA
| | | | - Motohiro Ebisawa
- />Department of Allergy, Clinical Research Center for Allergology and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - Rose Kamenwa
- />Department of Pediatrics and Child Health, Aga Khan University Hospital, Nairobi, Kenya
| | - Bee Wah Lee
- />Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Haiqi Li
- />Department of Primary Child Care, Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Susan Prescott
- />Department of Immunology, Perth Children’s Hospital, Telethon KIDS Institute, School of Paediatrics and Child Health, University of Western Australia, Crawley, Australia
| | - John J. Riva
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Family Medicine, McMaster University, Hamilton, ON Canada
| | - Lanny Rosenwasser
- />Allergy-Immunology Division, Children’s Mercy Hospital & University of Missouri – Kansas City School of Medicine, Kansas City, MO USA
| | - Hugh Sampson
- />Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Luigi Terracciano
- />Department of Child and Maternal Medicine, University of Milan Medical School at the Melloni Hospital, Milan, Italy
| | - Andrea Vereda
- />Allergology Department, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
| | - Susan Waserman
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Holger J. Schünemann
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Jan L. Brożek
- />Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, ON Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| |
Collapse
|
39
|
Parker D, Ahn D, Cohen T, Prince A. Innate Immune Signaling Activated by MDR Bacteria in the Airway. Physiol Rev 2016; 96:19-53. [PMID: 26582515 DOI: 10.1152/physrev.00009.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Health care-associated bacterial pneumonias due to multiple-drug resistant (MDR) pathogens are an important public health problem and are major causes of morbidity and mortality worldwide. In addition to antimicrobial resistance, these organisms have adapted to the milieu of the human airway and have acquired resistance to the innate immune clearance mechanisms that normally prevent pneumonia. Given the limited efficacy of antibiotics, bacterial clearance from the airway requires an effective immune response. Understanding how specific airway pathogens initiate and regulate innate immune signaling, and whether this response is excessive, leading to host-induced pathology may guide future immunomodulatory therapy. We will focus on three of the most important causes of health care-associated pneumonia, Staphylococcus aureus, Pseudomonas aeruginosa, and Klebsiella pneumoniae, and review the mechanisms through which an inappropriate or damaging innate immune response is stimulated, as well as describe how airway pathogens cause persistent infection by evading immune activation.
Collapse
Affiliation(s)
- Dane Parker
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Danielle Ahn
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Taylor Cohen
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Alice Prince
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| |
Collapse
|
40
|
Ozeki K, Furuta T, Asano M, Noda T, Nakamura M, Shibata Y, Okada E, Ojima T. Association of Hay Fever with the Failure of Helicobacter pylori Primary Eradication. Intern Med 2016; 55:1729-34. [PMID: 27374672 DOI: 10.2169/internalmedicine.55.6388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective Recently, the number of patients receiving Helicobacter pylori eradication treatment has dramatically increased in Japan, although the eradication rate has gradually decreased. Patient characteristics could affect the eradication rate. Our aim in this study was to investigate the association between failed first-line eradication therapy and hay fever. Methods We researched 356 patients who visited a pharmacy adjacent to the Internal Medicine clinic with a prescription for first-line H. pylori eradication treatment and investigated whether the patients had hay fever using a questionnaire. We separated these patients into 2 groups based on the success or failure of eradication according to the clinical data and performed a logistic regression analysis to investigate the influence of hay fever on first-line eradication failure. Results The eradication rate of patients with and without hay fever was 65.6% and 77.7%, respectively. The adjusted odds ratios according to which patients with hay fever would fail eradication therapy gradually lowered with increasing patient age [≤50 years, odds ratio (OR) 6.81, p=0.089; 51-60 years, OR 2.75, p=0.145; 61-70 years, OR 1.60, p=0.391; >70 years, OR 1.02, p=0.979]. A significant relationship was found for all patients (OR 1.88, p=0.047) and the age group ≤70 years (OR 2.31, p=0.024). Conclusion Patients with hay fever have difficulty with first-line eradication, especially younger patients. The existence of clarithromycin-resistant bacteria is suspected, and other factors may also be involved. When a hay fever sufferer receives first-line treatment, eradication might be difficult and other treatment may be required.
Collapse
Affiliation(s)
- Kayoko Ozeki
- Department of Community Health and Preventive Medicine, Hamamatsu University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Forsythe P. Microbes taming mast cells: Implications for allergic inflammation and beyond. Eur J Pharmacol 2015; 778:169-75. [PMID: 26130124 DOI: 10.1016/j.ejphar.2015.06.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/21/2015] [Accepted: 06/17/2015] [Indexed: 12/30/2022]
Abstract
There is increasing awareness of a relationship between our microbiota and the pathogenesis of allergy and other inflammatory diseases. In investigating the mechanisms underlying microbiota modulation of allergy the focus has been on the induction phase; alterations in the phenotype and function of antigen presenting cells, induction of regulatory T cells and shifts in Th1/Th2 balance. However there is evidence that microbes can influence the effector phase of disease, specifically that certain potentially beneficial bacteria can attenuate mast cell activation and degranulation. Furthermore, it appears that different non-pathogenic bacteria can utilize distinct mechanisms to stabilize mast cells, acting locally though direct interaction with the mast cell at mucosal sites or attenuating systemic mast cell dependent responses, likely through indirect signaling mechanisms. The position of mast cells on the frontline of defense against pathogens also suggests they may play an important role in fostering the host-microbiota relationship. Mast cells are also conduits of neuro-immuo-endocrine communication, suggesting the ability of microbes to modulate cell responses may have implications for host physiology beyond immunology. Further investigation of mast cell regulation by non-pathogenic or symbiotic bacteria will likely lead to a greater understanding of host microbiota interaction and the role of the microbiome in health and disease.
Collapse
Affiliation(s)
- Paul Forsythe
- McMaster Brain-Body Institute at St. Joseph's Healthcare, Hamilton, The Firestone Institute for Respiratory Health and Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
42
|
Lopez CA, Kingsbury DD, Velazquez EM, Bäumler AJ. Collateral damage: microbiota-derived metabolites and immune function in the antibiotic era. Cell Host Microbe 2015; 16:156-163. [PMID: 25121745 DOI: 10.1016/j.chom.2014.07.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our long-standing evolutionary association with gut-associated microbial communities has given rise to an intimate relationship, which affects many aspects of human health. Recent studies on the mechanisms that link these microbial communities to immune education, nutrition, and protection against pathogens point to microbiota-derived metabolites as key players during these microbe-host interactions. A disruption of gut-associated microbial communities by antibiotic treatment can result in a depletion of microbiota-derived metabolites, thereby enhancing pathogen susceptibility, impairing immune homeostasis, and contributing to the rise of certain chronic inflammatory diseases. Here, we highlight some of the recently elucidated mechanisms that showcase the impacts of microbiota-derived metabolites on human health.
Collapse
Affiliation(s)
- Christopher A Lopez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Dawn D Kingsbury
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Eric M Velazquez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
43
|
Seo JH, Kim HY, Jung YH, Lee E, Yang SI, Yu HS, Kim YJ, Kang MJ, Kim HJ, Park KS, Kwon JW, Kim BJ, Kim HB, Kim EJ, Lee JS, Lee SY, Hong SJ. Interactions between innate immunity genes and early-life risk factors in allergic rhinitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2015; 7:241-8. [PMID: 25840711 PMCID: PMC4397364 DOI: 10.4168/aair.2015.7.3.241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/25/2014] [Accepted: 07/29/2014] [Indexed: 12/12/2022]
Abstract
PURPOSE Allergic rhinitis (AR) is a common chronic disease. Many factors could affect the development of AR. We investigated early-life factors, such as delivery mode, feeding method, and use of antibiotics during infancy, which could affect the development of AR. In addition, how interactions between these factors and innate gene polymorphisms influence the development of AR was investigated. METHODS A cross-sectional study of 1,828 children aged 9-12 years was conducted. Three early-life factors and AR were assessed by a questionnaire. Skin prick tests were done. Polymorphisms of TLR4 (rs1927911) and CD14 (rs2569190) were genotyped. RESULTS Use of antibiotics during infancy increased the risk of AR (aOR [95% CI] 1.511 [1.222-2.037]) and atopic AR (aOR [95% CI], 1.565 [1.078-2.272]). There were synergistic interactions between caesarean delivery, formula feeding, and use of antibiotics in the rate of atopic AR (aOR [95% CI], 3.038 [1.256-7.347]). Additional analyses revealed that the risk for the development of AR or atopic AR subjects with the TLR4 CC genotype were highest when all the 3 early-life factors were present (aOR [95% CI], 5.127 [1.265-20.780] for AR; 6.078 [1.499-24.649] for atopic AR). In addition, the risk for the development of AR or atopic AR in subjects with the CD14 TT genotype were highest when all the 3 early-life factors were present (aOR [95% CI], 5.960 [1.421-15.002] for AR; 6.714 [1.440-31.312] for atopic AR). CONCLUSIONS Delivery mode, feeding method, and use of antibiotics during infancy appeared to have synergistic interactions in the development of AR. Gene-environment interactions between polymorphism of innate genes and early- life risk factors might affect the development of AR.
Collapse
Affiliation(s)
- Ju Hee Seo
- Department of Pediatrics, Korean Cancer Center Hospital, Seoul, Korea
| | - Hyung Young Kim
- Department of Pediatrics, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Young Ho Jung
- Department of Pediatrics, CHA University School of Medicine, Seongnam, Korea
| | - Eun Lee
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Research Center for Standardization of Allergic Diseases, University of Ulsan College of Medicine, Seoul, Korea
| | - Song I Yang
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Research Center for Standardization of Allergic Diseases, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho Sung Yu
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Young Joon Kim
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Mi Jin Kang
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Ha Jung Kim
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Kang Seo Park
- Department of Pediatrics, Presbyterian Medical Center, Jeonju, Korea
| | - Ji Won Kwon
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Byung Ju Kim
- Department of Pediatrics, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Hyo Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Eun Jin Kim
- Allergy TF, Department of Immunology and Pathology, Korea National Institute of Health, Osong, Korea
| | - Joo Shil Lee
- Allergy TF, Department of Immunology and Pathology, Korea National Institute of Health, Osong, Korea
| | - So Yeon Lee
- Department of Pediatrics, Hallym Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea.
| | - Soo Jong Hong
- Childhood Asthma Atopy Center, Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.; Research Center for Standardization of Allergic Diseases, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
44
|
Fiocchi A, Pawankar R, Cuello-Garcia C, Ahn K, Al-Hammadi S, Agarwal A, Beyer K, Burks W, Canonica GW, Ebisawa M, Gandhi S, Kamenwa R, Lee BW, Li H, Prescott S, Riva JJ, Rosenwasser L, Sampson H, Spigler M, Terracciano L, Vereda-Ortiz A, Waserman S, Yepes-Nuñez JJ, Brożek JL, Schünemann HJ. World Allergy Organization-McMaster University Guidelines for Allergic Disease Prevention (GLAD-P): Probiotics. World Allergy Organ J 2015; 8:4. [PMID: 25628773 PMCID: PMC4307749 DOI: 10.1186/s40413-015-0055-2] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Prevalence of allergic diseases in infants, whose parents and siblings do not have allergy, is approximately 10% and reaches 20-30% in those with an allergic first-degree relative. Intestinal microbiota may modulate immunologic and inflammatory systemic responses and, thus, influence development of sensitization and allergy. Probiotics have been reported to modulate immune responses and their supplementation has been proposed as a preventive intervention. OBJECTIVE The World Allergy Organization (WAO) convened a guideline panel to develop evidence-based recommendations about the use of probiotics in the prevention of allergy. METHODS We identified the most relevant clinical questions and performed a systematic review of randomized controlled trials of probiotics for the prevention of allergy. We followed the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach to develop recommendations. We searched for and reviewed the evidence about health effects, patient values and preferences, and resource use (up to November 2014). We followed the GRADE evidence-to-decision framework to develop recommendations. RESULTS Currently available evidence does not indicate that probiotic supplementation reduces the risk of developing allergy in children. However, considering all critical outcomes in this context, the WAO guideline panel determined that there is a likely net benefit from using probiotics resulting primarily from prevention of eczema. The WAO guideline panel suggests: a) using probiotics in pregnant women at high risk for having an allergic child; b) using probiotics in women who breastfeed infants at high risk of developing allergy; and c) using probiotics in infants at high risk of developing allergy. All recommendations are conditional and supported by very low quality evidence. CONCLUSIONS WAO recommendations about probiotic supplementation for prevention of allergy are intended to support parents, clinicians and other health care professionals in their decisions whether to use probiotics in pregnancy and during breastfeeding, and whether to give them to infants.
Collapse
Affiliation(s)
| | - Ruby Pawankar
- />Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Carlos Cuello-Garcia
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
- />Tecnologico de Monterrey School of Medicine and Health Sciences, Monterrey, Mexico
| | - Kangmo Ahn
- />Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Suleiman Al-Hammadi
- />Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Arnav Agarwal
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | | | - Wesley Burks
- />Department of Pediatrics, University of North Carolina, Chapel Hill, NC USA
| | | | - Motohiro Ebisawa
- />Department of Allergy, Clinical Research Center for Allergology and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - Shreyas Gandhi
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
- />Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Rose Kamenwa
- />Department of Pediatrics and Child Health, Aga Khan University Hospital, Nairobi, Kenya
| | - Bee Wah Lee
- />Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiqi Li
- />Department of Primary Child Care, Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Susan Prescott
- />Department of Immunology, Perth Children’s Hospital, Telethon KIDS Institute, School of Paediatrics and Child Health, University of Western Australia, Perth, Australia
| | - John J Riva
- />Department of Family Medicine, McMaster University, Hamilton, ON Canada
| | - Lanny Rosenwasser
- />Allergy-Immunology Division, Children’s Mercy Hospital & University of Missouri – Kansas City School of Medicine, Kansas City, MO USA
| | - Hugh Sampson
- />Jaffe Food Allergy Institute, Mount Sinai School of Medicine, New York, NY USA
| | | | - Luigi Terracciano
- />Department of Child and Maternal Medicine, University of Milan Medical School at the Melloni Hospital, Milan, Italy
| | - Andrea Vereda-Ortiz
- />Allergology Department, Hospital Infantil Universitario Niño Jesus, Madrid, Spain
| | - Susan Waserman
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Juan José Yepes-Nuñez
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
| | - Jan L Brożek
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| | - Holger J Schünemann
- />Department of Clinical Epidemiology and Biostatistics, McMaster University Health Sciences Centre, Room 2C19, 1200 Main Street West, Hamilton, ON L8N 3Z5 Canada
- />Department of Medicine, McMaster University, Hamilton, ON Canada
| |
Collapse
|
45
|
Beirão EM, Padovan ACB, Furtado JJD, Colombo AL, Medeiros EAS. Does the change on gastrointestinal tract microbiome affects host? Braz J Infect Dis 2014; 18:660-3. [PMID: 24835619 PMCID: PMC9425252 DOI: 10.1016/j.bjid.2014.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/28/2014] [Accepted: 04/21/2014] [Indexed: 01/04/2023] Open
Abstract
During the past decade, studies on the composition of human microbiota and its relation to the host became one of the most explored subjects of the medical literature. The development of high-throughput molecular technologies allowed a deeper characterization of human microbiota and a better understanding of its relationship with health and disease. Changes in human habits including wide use of antimicrobials can result in dysregulation of host–microbiome homeostasis, with multiple consequences. The purpose of this review is to highlight the most important evidence in the literature of host–microbiome interactions and illustrate how these intriguing relations may lead to new treatment and prevention strategies.
Collapse
Affiliation(s)
- Elisa M Beirão
- Department of Infectiology, Hospital Heliópolis, São Paulo, SP, Brazil.
| | - Ana Carolina B Padovan
- Micology Special Laboratory, Disciplina de Infectologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | | | - Arnaldo L Colombo
- Micology Special Laboratory, Disciplina de Infectologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Eduardo A S Medeiros
- Service of Hospital Infection Control, Disciplina de Infectologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
46
|
Arrieta MC, Stiemsma LT, Amenyogbe N, Brown EM, Finlay B. The intestinal microbiome in early life: health and disease. Front Immunol 2014; 5:427. [PMID: 25250028 PMCID: PMC4155789 DOI: 10.3389/fimmu.2014.00427] [Citation(s) in RCA: 607] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/22/2014] [Indexed: 12/27/2022] Open
Abstract
Human microbial colonization begins at birth and continues to develop and modulate in species abundance for about 3 years, until the microbiota becomes adult-like. During the same time period, children experience significant developmental changes that influence their health status as well as their immune system. An ever-expanding number of articles associate several diseases with early-life imbalances of the gut microbiota, also referred to as gut microbial dysbiosis. Whether early-life dysbiosis precedes and plays a role in disease pathogenesis, or simply originates from the disease process itself is a question that is beginning to be answered in a few diseases, including IBD, obesity, and asthma. This review describes the gut microbiome structure and function during the formative first years of life, as well as the environmental factors that determine its composition. It also aims to discuss the recent advances in understanding the role of the early-life gut microbiota in the development of immune-mediated, metabolic, and neurological diseases. A greater understanding of how the early-life gut microbiota impacts our immune development could potentially lead to novel microbial-derived therapies that target disease prevention at an early age.
Collapse
Affiliation(s)
- Marie-Claire Arrieta
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada
| | - Leah T Stiemsma
- Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | - Nelly Amenyogbe
- Child and Family Research Institute, University of British Columbia , Vancouver, BC , Canada
| | - Eric M Brown
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada
| | - Brett Finlay
- Michael Smith Laboratories, University of British Columbia , Vancouver, BC , Canada ; Department of Microbiology and Immunology, University of British Columbia , Vancouver, BC , Canada ; Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
47
|
Dranse HJ, Kelly MEM, Hudson BD. Drugs or diet?--Developing novel therapeutic strategies targeting the free fatty acid family of GPCRs. Br J Pharmacol 2014; 170:696-711. [PMID: 23937426 DOI: 10.1111/bph.12327] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 07/17/2013] [Accepted: 07/29/2013] [Indexed: 02/07/2023] Open
Abstract
Free fatty acids (FFAs) are metabolic intermediates that may be obtained through the diet, synthesized endogenously, or produced via fermentation of carbohydrates by gut microbiota. In addition to serving as an important source of energy, FFAs are known to produce a variety of both beneficial and detrimental effects on metabolic and inflammatory processes. While historically, FFAs were believed to produce these effects only through intracellular targets such as peroxisome proliferator-activated receptors, it has now become clear that FFAs are also agonists for several GPCRs, including a family of four receptors now termed FFA1-4. Increasing evidence suggests that FFA1-4 mediate many of the beneficial properties of FFAs and not surprisingly, this has generated significant interest in the potential of these receptors as therapeutic targets for the treatment of a variety of metabolic and inflammatory disorders. In addition to the traditional strategy of developing small-molecule therapeutics targeting these receptors, there has also been some consideration given to alternate therapeutic approaches, specifically by manipulating endogenous FFA concentrations through alteration of either dietary intake, or production by gut microbiota. In this review, the current state of knowledge for FFA1-4 will be discussed, together with their potential as therapeutic targets in the treatment of metabolic and inflammatory disorders. In particular, the evidence in support of small molecule versus dietary and microbiota-based therapeutic approaches will be considered to provide insight into the development of novel multifaceted strategies targeting the FFA receptors for the treatment of metabolic and inflammatory disorders.
Collapse
Affiliation(s)
- H J Dranse
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | | | | |
Collapse
|
48
|
El Aidy S, Dinan TG, Cryan JF. Immune modulation of the brain-gut-microbe axis. Front Microbiol 2014; 5:146. [PMID: 24778631 PMCID: PMC3985034 DOI: 10.3389/fmicb.2014.00146] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 03/20/2014] [Indexed: 12/11/2022] Open
Affiliation(s)
- Sahar El Aidy
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Industrial Biotechnology, Genetic Engineering and Biotechnology Research Institute, Sadat City University Sadat City, Egypt
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Psychiatry, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Anatomy and Neuroscience, University College Cork Cork, Ireland
| |
Collapse
|
49
|
Ong MS, Umetsu DT, Mandl KD. Consequences of antibiotics and infections in infancy: bugs, drugs, and wheezing. Ann Allergy Asthma Immunol 2014; 112:441-445.e1. [PMID: 24631182 DOI: 10.1016/j.anai.2014.01.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/05/2014] [Accepted: 01/26/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND The prevalence of asthma has increased alarmingly in the past 2 to 3 decades. Increased antibiotic use in infancy has been suggested to limit exposure to gastrointestinal microbes and to predispose to asthma in later life. OBJECTIVE To evaluate the association between antibiotic exposure during the first year of life and the development of asthma up to the age of 7 years. METHODS A retrospective population-based study of a cohort of children enrolled in a nationwide employer-provided health insurance plan from January 1, 1999, through December 31, 2006, in the United States (n = 62,576). We evaluated the association between antibiotic exposure during the first year of life and subsequent development of 3 asthma phenotypes: transient wheezing (began and resolved before 3 years of age), late-onset asthma (began after 3 years of age), and persistent asthma (began before 3 years of age and persisted through 4-7 years of age). RESULTS Antibiotic use in the first year of life was associated with the development of transient wheezing (odds ratio [OR], 2.0; 95% confidence interval [CI], 1.9-2.2; P < .001) and persistent asthma (OR, 1.6; 95% CI, 1.5-1.7; P < .001). A dose-response effect was observed. When 5 or more antibiotic courses were received, the odds of persistent asthma doubled (OR, 1.9; 95% CI, 1.5-2.6; P < .001). There is no association between antibiotic use and late-onset asthma. CONCLUSION Antibiotic use in the first year life is associated with an increased risk of early-onset childhood asthma that began before 3 years of age. The apparent effect has a clear dose response. Heightened caution about avoiding unnecessary use of antibiotics in infants is warranted.
Collapse
Affiliation(s)
- Mei-Sing Ong
- Australian Institute for Health Innovation, University of New South Wales, Sydney, Australia; Children's Hospital Informatics Program at Harvard-MIT Health Sciences and Technology, Boston Children's Hospital, Boston, Massachusetts
| | - Dale T Umetsu
- Division of Allergy and Immunology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Kenneth D Mandl
- Children's Hospital Informatics Program at Harvard-MIT Health Sciences and Technology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Center for Biomedical Informatics, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
50
|
Pfefferle PI, Renz H. Microbial exposure and onset of allergic diseases - potential prevention strategies? Allergol Int 2014; 63:3-10. [PMID: 24569150 DOI: 10.2332/allergolint.13-rai-0671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammatory diseases are a major health problem with global dimension. Particularly, the incidence of allergic diseases has been increased tremendously within the last decades. This world-wide trend clearly indicates the demand for new approaches in the investigation of early allergy development. Recent studies underlined the basic postulate of the hygiene hypothesis that early exposure to microbial stimuli plays a crucial role in the prevention of chronic inflammatory conditions in adulthood. There is ample evidence that, both, exogenous microbes and endogenous microbial communities, the human microbiota, shape the developing immune system and might be involved in prevention of pathologic pro-inflammatory trails. According to the Barker hypothesis, epidemiological studies pointed to transmaternal transmission from the mother to the offspring already in prenatal life. Experimental data from murine models support these findings. This state of the art review provides an overview on the current literature and presents new experimental concepts that point out to future application in the prevention of allergic diseases.
Collapse
Affiliation(s)
- Petra Ina Pfefferle
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics Philipps University Marburg, Biomedical Research Centre, Marburg, Germany; University of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Center for Lung Research (DZL), Marburg, Germany
| | - Harald Renz
- University of Gießen and Marburg Lung Center (UGMLC), Member of the German Lung Center for Lung Research (DZL), Marburg, Germany; Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics Philipps University Marburg, University Hospital Giessen and Marburg GmbH, Marburg, Germany
| |
Collapse
|